1
|
Sabnis SS, Narasimhan KKS, Chettiar PB, Gakare SG, Shelkar GP, Asati DG, Thakur SS, Dravid SM. Intravenous recombinant cerebellin 1 treatment restores signalling by spinal glutamate delta 1 receptors and mitigates chronic pain. Br J Pharmacol 2024; 181:1421-1437. [PMID: 38044332 PMCID: PMC11288346 DOI: 10.1111/bph.16296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 11/09/2023] [Accepted: 11/14/2023] [Indexed: 12/05/2023] Open
Abstract
BACKGROUND AND PURPOSE Chronic pain remains a major clinical problem that needs effective therapeutic agents. Glutamate delta 1 (GluD1) receptors and the protein cerebellin 1 (Cbln1) are down-regulated in the central amygdala (CeA) in models of inflammatory and neuropathic pain. One treatment with Cbln1, intracerebroventricularly (ICV) or in CeA, normalized GluD1 and reduced AMPA receptor expression, resulting in lasting (7-10 days) pain relief. Unlike many CNS-targeting biological agents, the structure of Cbln1 suggests potential blood-brain barrier penetration. Here, we have tested whether systemic administration of Cbln1 provides analgesic effects via action in the CNS. EXPERIMENTAL APPROACH Analgesic effects of intravenous recombinant Cbln1 was assessed in complete Freund's adjuvant inflammatory pain model in mice. GluD1 knockout and a mutant form of Cbln1 were used. KEY RESULTS A single intravenous injection of Cbln1 mitigated nocifensive and averse behaviour in both inflammatory and neuropathic pain models. This effect of Cbln1 was dependent on GluD1 receptors and required binding to the amino terminal domain of GluD1. Time course of analgesic effect was similar to previously reported ICV and intra-CeA injection. GluD1 in both spinal cord and CeA was down -regulated in the inflammatory pain model, whereas GluD1 expression in spinal cord but not in CeA, was partly normalized by intravenous Cbln1. Importantly, recombinant Cbln1 was detected in the synaptoneurosomes in spinal cord but not in the CeA. CONCLUSIONS AND IMPLICATIONS Our results describe a novel mechanism by which systemic Cbln1 induces analgesia potentially by central actions involving normalization of signalling by spinal cord GluD1 receptors.
Collapse
Affiliation(s)
- Siddhesh S. Sabnis
- Department of Pharmacology and Neuroscience, Creighton University, 2500 California Plaza, Omaha, Nebraska 68178, USA
| | - Kishore Kumar S. Narasimhan
- Department of Pharmacology and Neuroscience, Creighton University, 2500 California Plaza, Omaha, Nebraska 68178, USA
| | - Poojashree B. Chettiar
- Department of Pharmacology and Neuroscience, Creighton University, 2500 California Plaza, Omaha, Nebraska 68178, USA
| | - Sukanya G. Gakare
- Department of Pharmacology and Neuroscience, Creighton University, 2500 California Plaza, Omaha, Nebraska 68178, USA
| | - Gajanan P. Shelkar
- Department of Pharmacology and Neuroscience, Creighton University, 2500 California Plaza, Omaha, Nebraska 68178, USA
| | - Devansh G. Asati
- Department of Pharmacology and Neuroscience, Creighton University, 2500 California Plaza, Omaha, Nebraska 68178, USA
| | - Shriti S. Thakur
- Department of Pharmacology and Neuroscience, Creighton University, 2500 California Plaza, Omaha, Nebraska 68178, USA
| | - Shashank M. Dravid
- Department of Pharmacology and Neuroscience, Creighton University, 2500 California Plaza, Omaha, Nebraska 68178, USA
| |
Collapse
|
2
|
Wang M, Caryotakis SE, Smith GG, Nguyen AV, Pleasure DE, Soulika AM. CSF1R antagonism results in increased supraspinal infiltration in EAE. J Neuroinflammation 2024; 21:103. [PMID: 38643194 PMCID: PMC11031888 DOI: 10.1186/s12974-024-03063-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Accepted: 03/11/2024] [Indexed: 04/22/2024] Open
Abstract
BACKGROUND Colony stimulating factor 1 receptor (CSF1R) signaling is crucial for the maintenance and function of various myeloid subsets. CSF1R antagonism was previously shown to mitigate clinical severity in experimental autoimmune encephalomyelitis (EAE). The associated mechanisms are still not well delineated. METHODS To assess the effect of CSF1R signaling, we employed the CSF1R antagonist PLX5622 formulated in chow (PLX5622 diet, PD) and its control chow (control diet, CD). We examined the effect of PD in steady state and EAE by analyzing cells isolated from peripheral immune organs and from the CNS via flow cytometry. We determined CNS infiltration sites and assessed the extent of demyelination using immunohistochemistry of cerebella and spinal cords. Transcripts of genes associated with neuroinflammation were also analyzed in these tissues. RESULTS In addition to microglial depletion, PD treatment reduced dendritic cells and macrophages in peripheral immune organs, both during steady state and during EAE. Furthermore, CSF1R antagonism modulated numbers and relative frequencies of T effector cells both in the periphery and in the CNS during the early stages of the disease. Classical neurological symptoms were milder in PD compared to CD mice. Interestingly, a subset of PD mice developed atypical EAE symptoms. Unlike previous studies, we observed that the CNS of PD mice was infiltrated by increased numbers of peripheral immune cells compared to that of CD mice. Immunohistochemical analysis showed that CNS infiltrates in PD mice were mainly localized in the cerebellum while in CD mice infiltrates were primarily localized in the spinal cords during the onset of neurological deficits. Accordingly, during the same timepoint, cerebella of PD but not of CD mice had extensive demyelinating lesions, while spinal cords of CD but not of PD mice were heavily demyelinated. CONCLUSIONS Our findings suggest that CSF1R activity modulates the cellular composition of immune cells both in the periphery and within the CNS, and affects lesion localization during the early EAE stages.
Collapse
Affiliation(s)
- Marilyn Wang
- Department of Dermatology, School of Medicine, University of California, Davis, Sacramento, CA, USA
| | - Sofia E Caryotakis
- Shriners Hospitals for Children, Northern California, Sacramento, CA, USA
- University of California, San Francisco, San Francisco, CA, USA
| | - Glendalyn G Smith
- Shriners Hospitals for Children, Northern California, Sacramento, CA, USA
| | - Alan V Nguyen
- Department of Dermatology, School of Medicine, University of California, Davis, Sacramento, CA, USA
- Sutro Biosciences, South San Francisco, CA, USA
| | - David E Pleasure
- Shriners Hospitals for Children, Northern California, Sacramento, CA, USA
- Department of Neurology, School of Medicine, University of California, Davis, Sacramento, CA, USA
| | - Athena M Soulika
- Department of Dermatology, School of Medicine, University of California, Davis, Sacramento, CA, USA.
- Shriners Hospitals for Children, Northern California, Sacramento, CA, USA.
| |
Collapse
|
3
|
Lu L, Shi Y, Wei B, Li W, Yu X, Zhao Y, Yu D, Sun M. YTHDF3 modulates the Cbln1 level by recruiting BTG2 and is implicated in the impaired cognition of prenatal hypoxia offspring. iScience 2024; 27:108703. [PMID: 38205248 PMCID: PMC10776956 DOI: 10.1016/j.isci.2023.108703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 09/22/2023] [Accepted: 12/06/2023] [Indexed: 01/12/2024] Open
Abstract
The "Fetal Origins of Adult Disease (FOAD)" hypothesis holds that adverse factors during pregnancy can increase the risk of chronic diseases in offspring. Here, we investigated the effects of prenatal hypoxia (PH) on brain structure and function in adult offspring and explored the role of the N6-methyladenosine (m6A) pathway. The results suggest that abnormal cognition in PH offspring may be related to the dysregulation of the m6A pathway, specifically increased levels of YTHDF3 in the hippocampus. YTHDF3 interacts with BTG2 and is involved in the decay of Cbln1 mRNA, leading to the down-regulation of Cbln1 expression. Deficiency of Cbln1 may contribute to abnormal synaptic function, which in turn causes cognitive impairment in PH offspring. This study provides a scientific clues for understanding the mechanisms of impaired cognition in PH offspring and provides a theoretical basis for the treatment of cognitive impairment in offspring exposed to PH.
Collapse
Affiliation(s)
- Likui Lu
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou City, Jiangsu, China
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yajun Shi
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou City, Jiangsu, China
| | - Bin Wei
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou City, Jiangsu, China
| | - Weisheng Li
- Department of Gynaecology, Qingdao Hospital, University of Health and Rehabilitation Sciences (Qingdao Municipal Hospital), Qingdao, Shandong, China
| | - Xi Yu
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou City, Jiangsu, China
| | - Yan Zhao
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou City, Jiangsu, China
| | - Dongyi Yu
- Center for Medical Genetics and Prenatal Diagnosis, Key Laboratory of Birth Defect Prevention and Genetic, Medicine of Shandong Health Commission, Key Laboratory of Birth Regulation and Control Technology of National Health Commission of China, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, Shandong, China
| | - Miao Sun
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou City, Jiangsu, China
- Dushu Lake Hospital Affiliated to Soochow University, Suzhou, Jiangsu Province, China
| |
Collapse
|
4
|
Rudolph S, Badura A, Lutzu S, Pathak SS, Thieme A, Verpeut JL, Wagner MJ, Yang YM, Fioravante D. Cognitive-Affective Functions of the Cerebellum. J Neurosci 2023; 43:7554-7564. [PMID: 37940582 PMCID: PMC10634583 DOI: 10.1523/jneurosci.1451-23.2023] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 08/21/2023] [Accepted: 08/22/2023] [Indexed: 11/10/2023] Open
Abstract
The cerebellum, traditionally associated with motor coordination and balance, also plays a crucial role in various aspects of higher-order function and dysfunction. Emerging research has shed light on the cerebellum's broader contributions to cognitive, emotional, and reward processes. The cerebellum's influence on autonomic function further highlights its significance in regulating motivational and emotional states. Perturbations in cerebellar development and function have been implicated in various neurodevelopmental disorders, including autism spectrum disorder and attention deficit hyperactivity disorder. An increasing appreciation for neuropsychiatric symptoms that arise from cerebellar dysfunction underscores the importance of elucidating the circuit mechanisms that underlie complex interactions between the cerebellum and other brain regions for a comprehensive understanding of complex behavior. By briefly discussing new advances in mapping cerebellar function in affective, cognitive, autonomic, and social processing and reviewing the role of the cerebellum in neuropathology beyond the motor domain, this Mini-Symposium review aims to provide a broad perspective of cerebellar intersections with the limbic brain in health and disease.
Collapse
Affiliation(s)
- Stephanie Rudolph
- Department of Neuroscience, Department of Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, New York, New York 10461
| | - Aleksandra Badura
- Department of Neuroscience, Erasmus MC Rotterdam, Rotterdam, 3015 GD, The Netherlands
| | - Stefano Lutzu
- Department of Neuroscience, Department of Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, New York, New York 10461
| | - Salil Saurav Pathak
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, Minnesota 55812
| | - Andreas Thieme
- Department of Neurology and Center for Translational Neuro- and Behavioral Sciences, University Hospital Essen, Essen, D-45147, Germany
| | - Jessica L Verpeut
- Department of Psychology, Arizona State University, Tempe, Arizona 85287
| | - Mark J Wagner
- National Institute of Neurological Disorders & Stroke, National Institutes of Health, Bethesda, Maryland 20814
| | - Yi-Mei Yang
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, Minnesota 55812
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota 55455
| | - Diasynou Fioravante
- Center for Neuroscience, University of California-Davis, Davis, California 95618
- Department of Neurobiology, Physiology and Behavior, University of California-Davis, Davis, California 95618
| |
Collapse
|
5
|
Chen Q, Xu Y, Christiaen E, Wu GR, De Witte S, Vanhove C, Saunders J, Peremans K, Baeken C. Structural connectome alterations in anxious dogs: a DTI-based study. Sci Rep 2023; 13:9946. [PMID: 37337053 DOI: 10.1038/s41598-023-37121-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 06/15/2023] [Indexed: 06/21/2023] Open
Abstract
Anxiety and fear are dysfunctional behaviors commonly observed in domesticated dogs. Although dogs and humans share psychopathological similarities, little is known about how dysfunctional fear behaviors are represented in brain networks in dogs diagnosed with anxiety disorders. A combination of diffusion tensor imaging (DTI) and graph theory was used to investigate the underlying structural connections of dysfunctional anxiety in anxious dogs and compared with healthy dogs with normal behavior. The degree of anxiety was assessed using the Canine Behavioral Assessment & Research Questionnaire (C-BARQ), a widely used, validated questionnaire for abnormal behaviors in dogs. Anxious dogs showed significantly decreased clustering coefficient ([Formula: see text]), decreased global efficiency ([Formula: see text]), and increased small-worldness (σ) when compared with healthy dogs. The nodal parameters that differed between the anxious dogs and healthy dogs were mainly located in the posterior part of the brain, including the occipital lobe, posterior cingulate gyrus, hippocampus, mesencephalon, and cerebellum. Furthermore, the nodal degree ([Formula: see text]) of the left cerebellum was significantly negatively correlated with "excitability" in the C-BARQ of anxious dogs. These findings could contribute to the understanding of a disrupted brain structural connectome underlying the pathological mechanisms of anxiety-related disorders in dogs.
Collapse
Affiliation(s)
- Qinyuan Chen
- Ghent Experimental Psychiatry (GHEP) Lab, Department of Head and Skin, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium.
| | - Yangfeng Xu
- Ghent Experimental Psychiatry (GHEP) Lab, Department of Head and Skin, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
- Department of Morphology, Imaging, Orthopedics, Rehabilitation and Nutrition, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Emma Christiaen
- Medical Image and Signal Processing (MEDISIP), Department of Electronics and Information Systems, Faculty of Engineering and Architecture, Ghent University, Ghent, Belgium
| | - Guo-Rong Wu
- Key Laboratory of Cognition and Personality, Faculty of Psychology, Southwest University, Chongqing, China
- School of Psychology, Jiangxi Normal University, Nanchang, China
| | - Sara De Witte
- Ghent Experimental Psychiatry (GHEP) Lab, Department of Head and Skin, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
- Department of Neurology and Bru-BRAIN, University Hospital (UZ Brussel), Brussels, Belgium
- Neuroprotection & Neuromodulation Research Group (NEUR), Center for Neurosciences (C4N), Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Christian Vanhove
- Medical Image and Signal Processing (MEDISIP), Department of Electronics and Information Systems, Faculty of Engineering and Architecture, Ghent University, Ghent, Belgium
| | - Jimmy Saunders
- Department of Morphology, Imaging, Orthopedics, Rehabilitation and Nutrition, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Kathelijne Peremans
- Department of Morphology, Imaging, Orthopedics, Rehabilitation and Nutrition, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Chris Baeken
- Ghent Experimental Psychiatry (GHEP) Lab, Department of Head and Skin, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
- Vrije Universiteit Brussel (VUB), Department of Psychiatry, University Hospital (UZ Brussel), Brussels, Belgium
- Department of Electrical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
| |
Collapse
|
6
|
Lin PY, Chen LY, Zhou P, Lee SJ, Trotter JH, Südhof TC. Neurexin-2 restricts synapse numbers and restrains the presynaptic release probability by an alternative splicing-dependent mechanism. Proc Natl Acad Sci U S A 2023; 120:e2300363120. [PMID: 36961922 PMCID: PMC10068831 DOI: 10.1073/pnas.2300363120] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 02/21/2023] [Indexed: 03/26/2023] Open
Abstract
α- and β-neurexins are extensively alternatively spliced, presynaptic cell-adhesion molecules that are thought to organize synapse assembly. However, recent data revealed that, in the hippocampus in vivo, the deletion of one neurexin isoform, Nrxn2, surprisingly increased excitatory synapse numbers and enhanced their presynaptic release probability, suggesting that Nrxn2 restricts, instead of enabling, synapse assembly. To delineate the synaptic function and mechanism of action of Nrxn2, we examined cultured hippocampal neurons as a reduced system. In heterologous synapse formation assays, different alternatively spliced Nrxn2β isoforms robustly promoted synapse assembly similar to Nrxn1β and Nrxn3β, consistent with a general synaptogenic function of neurexins. Deletion of Nrxn2 from cultured hippocampal neurons, however, caused a significant increase in synapse density and release probability, replicating the in vivo data that suggested a synapse-restricting function. Rescue experiments revealed that two of the four Nrxn2β splice variants (Nrxn2β-SS4+/SS5- and Nrxn2β-SS4+/SS5+) reversed the increase in synapse density in Nrxn2-deficient neurons, whereas only one of the four Nrxn2β splice variants (Nrxn2β-SS4+/SS5+) normalized the increase in release probability in Nrxn2-deficient neurons. Thus, a subset of Nrxn2 splice variants restricts synapse numbers and restrains their release probability in cultured neurons.
Collapse
Affiliation(s)
- Pei-Yi Lin
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA94305
| | - Lulu Y. Chen
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA94305
| | - Peng Zhou
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA94305
- HHMI, Stanford University School of Medicine, Stanford, CA94305
| | - Sung-Jin Lee
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA94305
| | - Justin H. Trotter
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA94305
| | - Thomas C. Südhof
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA94305
- HHMI, Stanford University School of Medicine, Stanford, CA94305
| |
Collapse
|
7
|
Song JHT, Ruven C, Patel P, Ding F, Macklis JD, Sahni V. Cbln1 Directs Axon Targeting by Corticospinal Neurons Specifically toward Thoraco-Lumbar Spinal Cord. J Neurosci 2023; 43:1871-1887. [PMID: 36823038 PMCID: PMC10027075 DOI: 10.1523/jneurosci.0710-22.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Revised: 01/24/2023] [Accepted: 02/02/2023] [Indexed: 02/25/2023] Open
Abstract
Corticospinal neurons (CSN) are centrally required for skilled voluntary movement, which necessitates that they establish precise subcerebral connectivity with the brainstem and spinal cord. However, molecular controls regulating specificity of this projection targeting remain largely unknown. We previously identified that developing CSN subpopulations exhibit striking axon targeting specificity in the spinal white matter. These CSN subpopulations with segmentally distinct spinal projections are also molecularly distinct; a subset of differentially expressed genes between these distinct CSN subpopulations regulate differential axon projection targeting. Rostrolateral CSN extend axons exclusively to bulbar-cervical segments (CSNBC-lat), while caudomedial CSN (CSNmedial) are more heterogeneous, with distinct, intermingled subpopulations extending axons to either bulbar-cervical or thoraco-lumbar segments. Here, we report, in male and female mice, that Cerebellin 1 (Cbln1) is expressed specifically by CSN in medial, but not lateral, sensorimotor cortex. Cbln1 shows highly dynamic temporal expression, with Cbln1 levels in CSN highest during the period of peak axon extension toward thoraco-lumbar segments. Using gain-of-function experiments, we identify that Cbln1 is sufficient to direct thoraco-lumbar axon extension by CSN. Misexpression of Cbln1 in CSNBC-lat either by in utero electroporation, or by postmitotic AAV-mediated gene delivery, redirects these axons past their normal bulbar-cervical targets toward thoracic segments. Further, Cbln1 overexpression in postmitotic CSNBC-lat increases the number of CSNmedial axons that extend past cervical segments into the thoracic cord. Collectively, these results identify that Cbln1 functions as a potent molecular control over thoraco-lumbar CSN axon extension, part of an integrated network of controls over segmentally-specific CSN axon projection targeting.SIGNIFICANCE STATEMENT Corticospinal neurons (CSN) exhibit remarkable diversity and precision of axonal projections to targets in the brainstem and distinct spinal segments; the molecular basis for this targeting diversity is largely unknown. CSN subpopulations projecting to distinct targets are also molecularly distinguishable. Distinct subpopulations degenerate in specific motor neuron diseases, further suggesting that intrinsic molecular differences might underlie differential vulnerability to disease. Here, we identify a novel molecular control, Cbln1, expressed by CSN extending axons to thoraco-lumbar spinal segments. Cbln1 is sufficient, but not required, for CSN axon extension toward distal spinal segments, and Cbln1 expression is controlled by recently identified, CSN-intrinsic regulators of axon extension. Our results identify that Cbln1, together with other regulators, coordinates segmentally precise CSN axon targeting.
Collapse
Affiliation(s)
- Janet H T Song
- Department of Stem Cell and Regenerative Biology, and Center for Brain Science, Harvard University, Cambridge, Massachusetts 02138
| | - Carolin Ruven
- Burke Neurological Institute, White Plains, New York 10605
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, New York 10065
| | - Payal Patel
- Burke Neurological Institute, White Plains, New York 10605
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, New York 10065
| | - Frances Ding
- Department of Stem Cell and Regenerative Biology, and Center for Brain Science, Harvard University, Cambridge, Massachusetts 02138
| | - Jeffrey D Macklis
- Department of Stem Cell and Regenerative Biology, and Center for Brain Science, Harvard University, Cambridge, Massachusetts 02138
| | - Vibhu Sahni
- Department of Stem Cell and Regenerative Biology, and Center for Brain Science, Harvard University, Cambridge, Massachusetts 02138
- Burke Neurological Institute, White Plains, New York 10605
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, New York 10065
| |
Collapse
|
8
|
Wang M, Tutt JO, Dorricott NO, Parker KL, Russo AF, Sowers LP. Involvement of the cerebellum in migraine. Front Syst Neurosci 2022; 16:984406. [PMID: 36313527 PMCID: PMC9608746 DOI: 10.3389/fnsys.2022.984406] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Accepted: 09/27/2022] [Indexed: 11/14/2022] Open
Abstract
Migraine is a disabling neurological disease characterized by moderate or severe headaches and accompanied by sensory abnormalities, e.g., photophobia, allodynia, and vertigo. It affects approximately 15% of people worldwide. Despite advancements in current migraine therapeutics, mechanisms underlying migraine remain elusive. Within the central nervous system, studies have hinted that the cerebellum may play an important sensory integrative role in migraine. More specifically, the cerebellum has been proposed to modulate pain processing, and imaging studies have revealed cerebellar alterations in migraine patients. This review aims to summarize the clinical and preclinical studies that link the cerebellum to migraine. We will first discuss cerebellar roles in pain modulation, including cerebellar neuronal connections with pain-related brain regions. Next, we will review cerebellar symptoms and cerebellar imaging data in migraine patients. Lastly, we will highlight the possible roles of the neuropeptide calcitonin gene-related peptide (CGRP) in migraine symptoms, including preclinical cerebellar studies in animal models of migraine.
Collapse
Affiliation(s)
- Mengya Wang
- Department of Neuroscience and Pharmacology, University of Iowa, Iowa City, IA, United States
| | - Joseph O. Tutt
- Department of Biology and Biochemistry, University of Bath, Bath, United Kingdom
| | | | - Krystal L. Parker
- Department of Psychiatry, University of Iowa, Iowa City, IA, United States
| | - Andrew F. Russo
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, IA, United States,Department of Neurology, University of Iowa, Iowa City, IA, United States,Center for the Prevention and Treatment of Visual Loss, Veterans Administration Health Center, Iowa City, IA, United States
| | - Levi P. Sowers
- Center for the Prevention and Treatment of Visual Loss, Veterans Administration Health Center, Iowa City, IA, United States,Department of Pediatrics, University of Iowa, Iowa City, IA, United States,*Correspondence: Levi P. Sowers
| |
Collapse
|
9
|
Dai J, Liakath-Ali K, Golf SR, Südhof TC. Distinct neurexin-cerebellin complexes control AMPA- and NMDA-receptor responses in a circuit-dependent manner. eLife 2022; 11:e78649. [PMID: 36205393 PMCID: PMC9586558 DOI: 10.7554/elife.78649] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 10/06/2022] [Indexed: 01/11/2023] Open
Abstract
At CA1→subiculum synapses, alternatively spliced neurexin-1 (Nrxn1SS4+) and neurexin-3 (Nrxn3SS4+) enhance NMDA-receptors and suppress AMPA-receptors, respectively, without affecting synapse formation. Nrxn1SS4+ and Nrxn3SS4+ act by binding to secreted cerebellin-2 (Cbln2) that in turn activates postsynaptic GluD1 receptors. Whether neurexin-Cbln2-GluD1 signaling has additional functions besides regulating NMDA- and AMPA-receptors, and whether such signaling performs similar roles at other synapses, however, remains unknown. Here, we demonstrate using constitutive Cbln2 deletions in mice that at CA1→subiculum synapses, Cbln2 performs no additional developmental roles besides regulating AMPA- and NMDA-receptors. Moreover, low-level expression of functionally redundant Cbln1 did not compensate for a possible synapse-formation function of Cbln2 at CA1→subiculum synapses. In exploring the generality of these findings, we examined the prefrontal cortex where Cbln2 was recently implicated in spinogenesis, and the cerebellum where Cbln1 is known to regulate parallel-fiber synapses. In the prefrontal cortex, Nrxn1SS4+-Cbln2 signaling selectively controlled NMDA-receptors without affecting spine or synapse numbers, whereas Nrxn3SS4+-Cbln2 signaling had no apparent role. In the cerebellum, conversely, Nrxn3SS4+-Cbln1 signaling regulated AMPA-receptors, whereas now Nrxn1SS4+-Cbln1 signaling had no manifest effect. Thus, Nrxn1SS4+- and Nrxn3SS4+-Cbln1/2 signaling complexes differentially control NMDA- and AMPA-receptors in different synapses in diverse neural circuits without regulating synapse or spine formation.
Collapse
Affiliation(s)
- Jinye Dai
- Howard Hughes Medical Institute, Stanford UniversityStanfordUnited States
- Department of Molecular and Cellular Physiology, Stanford UniversityStanfordUnited States
| | - Kif Liakath-Ali
- Department of Molecular and Cellular Physiology, Stanford UniversityStanfordUnited States
| | - Samantha Rose Golf
- Department of Molecular and Cellular Physiology, Stanford UniversityStanfordUnited States
| | - Thomas C Südhof
- Howard Hughes Medical Institute, Stanford UniversityStanfordUnited States
- Department of Molecular and Cellular Physiology, Stanford UniversityStanfordUnited States
| |
Collapse
|
10
|
GÜLER Ö, TUĞAN YILDIZ B, HAKKOYMAZ H, AYDIN S, YARDIM M. Levels of Serum and Urine Catecholaminergic and Apelinergic System Members in Acute Ischemic Stroke Patients. KAHRAMANMARAŞ SÜTÇÜ İMAM ÜNIVERSITESI TIP FAKÜLTESI DERGISI 2022. [DOI: 10.17517/ksutfd.1168625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/31/2023] Open
Abstract
Objective: To compare levels of catecholaminergic system members, renalase, cerebellin, and their substrates, epinephrine, norepinephrine, and dopamine, and apelinergic system members, apelin, elabela, and nitric oxide in the blood and urine of patients with acute ischemic stroke and healthy controls.
Materials and Methods: 42 patients with acute ischemic stroke and 42 age and sex-matched healthy controls were included in the study. Blood and urine samples were collected simultaneously and within the first 24 hours after the onset of acute stroke clinical manifestations and were measured using an ELISA method.
Results: The levels of serum and urine cerebellin, renalase, epinephrine, norepinephrine, dopamine, apelin, elebela, and nitric oxide were similar in ischemic stroke and in control groups (P>0.05). Strong correlations were found between renalase, cerebellin, and catecholamine levels in serum and urine (p
Collapse
|
11
|
Nozawa K, Sogabe T, Hayashi A, Motohashi J, Miura E, Arai I, Yuzaki M. In vivo nanoscopic landscape of neurexin ligands underlying anterograde synapse specification. Neuron 2022; 110:3168-3185.e8. [PMID: 36007521 DOI: 10.1016/j.neuron.2022.07.027] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 05/04/2022] [Accepted: 07/27/2022] [Indexed: 11/17/2022]
Abstract
Excitatory synapses are formed and matured by the cooperative actions of synaptic organizers, such as neurexins (Nrxns), neuroligins (Nlgns), LRRTMs, and Cbln1. Recent super-resolution nanoscopy developments have revealed that many synaptic organizers, as well as glutamate receptors and glutamate release machinery, exist as nanoclusters within synapses. However, it is unclear how such nanodomains interact with each other to organize excitatory synapses in vivo. By applying X10 expansion microscopy to epitope tag knockin mice, we found that Cbln1, Nlgn1, and LRRTM1, which share Nrxn as a common presynaptic receptor, form overlapping or separate nanodomains depending on Nrxn with or without a sequence encoded by splice site 4. The size and position of glutamate receptor nanodomains of GluD1, NMDA, and AMPA receptors were regulated by Cbln1, Nlgn1, and LRRTM1 nanodomains, respectively. These findings indicate that Nrxns anterogradely regulate the postsynaptic nanoscopic architecture of glutamate receptors through competition and coordination of Nrxn ligands.
Collapse
Affiliation(s)
- Kazuya Nozawa
- Department of Physiology, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Taku Sogabe
- Department of Physiology, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Ayumi Hayashi
- Department of Physiology, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Junko Motohashi
- Department of Physiology, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Eriko Miura
- Department of Physiology, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Itaru Arai
- Department of Physiology, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Michisuke Yuzaki
- Department of Physiology, Keio University School of Medicine, Tokyo 160-8582, Japan.
| |
Collapse
|
12
|
Hua JC, Xu XM, Xu ZG, Xue Y, Xu JJ, Hu JH, Wu Y, Chen YC. Abnormal cerebellar network and effective connectivity in sudden and long-term sensorineural hearing loss. Front Aging Neurosci 2022; 14:964349. [PMID: 36034151 PMCID: PMC9403534 DOI: 10.3389/fnagi.2022.964349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 07/14/2022] [Indexed: 11/17/2022] Open
Abstract
Sudden sensorineural hearing loss (SSNHL) is a common otology emergency and some SSNHL will develop into a long-term hearing loss (LSNHL). However, whether SSNHL and LSNHL have similar psychiatric patterns remains unknown, as well as the neural substrates. Increasing evidence has proved that the cerebellar network plays a vital role in hearing, cognition processing, and emotion control. Thus, we recruited 20 right SSNHL (RSSNHL), 20 right LSNHL (RLSNHL), and 24 well-matched healthy controls to explore the cerebellar patterns among the three groups. Every participant underwent pure tone audiometry tests, neuropsychological evaluations, and MRI scanning. Independent component analysis (ICA) was carried out on the MRI data and the cerebellar network was extracted. Granger causality analysis (GCA) was conducted using the significant cerebellar region as a seed. Pearson’s correlation analysis was computed between imaging characteristics and clinical features. ICA found the effect of group on right cerebellum lobule V for the cerebellar network. Then, we found decreased outflow from right cerebellum lobule V to right middle orbitofrontal cortex, inferior frontal gyrus, anterior cingulate cortex, superior temporal gyrus, and dorsal lateral prefrontal cortex in RSSNHL group in GCA analysis. No significance was found in RLSNHL subjects. Additionally, the RSSNHL group showed increased effective connectivity from the right middle frontal gyrus (MFG) and the RLSNHL group showed increased effective connectivity from the right insula and temporal pole to the right cerebellum lobule V. Moreover, connections between right cerebellum lobule V and mean time series of the cerebellar network was negatively correlated with anxiety score in RSSNHL and negatively correlated with depression scores in RLSNHL. Effective connectivity from right MFG to right cerebellum lobule V could predict anxiety status in RSSNHL subjects. Our results may prove potential imaging biomarkers and treatment targets for hearing loss in future work.
Collapse
Affiliation(s)
- Jin-Chao Hua
- Department of Otolaryngology, Nanjing Pukou Central Hospital, Pukou Branch Hospital of Jiangsu Province Hospital, Nanjing, China
| | - Xiao-Min Xu
- Department of Radiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Zhen-Gui Xu
- Department of Otolaryngology, Nanjing Pukou Central Hospital, Pukou Branch Hospital of Jiangsu Province Hospital, Nanjing, China
| | - Yuan Xue
- Department of Otolaryngology, Nanjing Pukou Central Hospital, Pukou Branch Hospital of Jiangsu Province Hospital, Nanjing, China
| | - Jin-Jing Xu
- Department of Otolaryngology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Jing-Hua Hu
- Department of Otolaryngology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Yuanqing Wu
- Department of Otolaryngology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
- *Correspondence: Yu-Chen Chen Yuanqing Wu
| | - Yu-Chen Chen
- Department of Radiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
- *Correspondence: Yu-Chen Chen Yuanqing Wu
| |
Collapse
|
13
|
Jung SJ, Vlasov K, D’Ambra AF, Parigi A, Baya M, Frez EP, Villalobos J, Fernandez-Frentzel M, Anguiano M, Ideguchi Y, Antzoulatos EG, Fioravante D. Novel Cerebello-Amygdala Connections Provide Missing Link Between Cerebellum and Limbic System. Front Syst Neurosci 2022; 16:879634. [PMID: 35645738 PMCID: PMC9136059 DOI: 10.3389/fnsys.2022.879634] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Accepted: 04/05/2022] [Indexed: 02/06/2023] Open
Abstract
The cerebellum is emerging as a powerful regulator of cognitive and affective processing and memory in both humans and animals and has been implicated in affective disorders. How the cerebellum supports affective function remains poorly understood. The short-latency (just a few milliseconds) functional connections that were identified between the cerebellum and amygdala—a structure crucial for the processing of emotion and valence—more than four decades ago raise the exciting, yet untested, possibility that a cerebellum-amygdala pathway communicates information important for emotion. The major hurdle in rigorously testing this possibility is the lack of knowledge about the anatomy and functional connectivity of this pathway. Our initial anatomical tracing studies in mice excluded the existence of a direct monosynaptic connection between the cerebellum and amygdala. Using transneuronal tracing techniques, we have identified a novel disynaptic circuit between the cerebellar output nuclei and the basolateral amygdala. This circuit recruits the understudied intralaminar thalamus as a node. Using ex vivo optophysiology and super-resolution microscopy, we provide the first evidence for the functionality of the pathway, thus offering a missing mechanistic link between the cerebellum and amygdala. This discovery provides a connectivity blueprint between the cerebellum and a key structure of the limbic system. As such, it is the requisite first step toward obtaining new knowledge about cerebellar function in emotion, thus fundamentally advancing understanding of the neurobiology of emotion, which is perturbed in mental and autism spectrum disorders.
Collapse
Affiliation(s)
- Se Jung Jung
- Center for Neuroscience, University of California, Davis, Davis, CA, United States
| | - Ksenia Vlasov
- Center for Neuroscience, University of California, Davis, Davis, CA, United States
| | - Alexa F. D’Ambra
- Center for Neuroscience, University of California, Davis, Davis, CA, United States
| | - Abhijna Parigi
- Center for Neuroscience, University of California, Davis, Davis, CA, United States
| | - Mihir Baya
- Center for Neuroscience, University of California, Davis, Davis, CA, United States
| | - Edbertt Paul Frez
- Center for Neuroscience, University of California, Davis, Davis, CA, United States
| | | | | | - Maribel Anguiano
- Center for Neuroscience, University of California, Davis, Davis, CA, United States
| | - Yoichiro Ideguchi
- Center for Neuroscience, University of California, Davis, Davis, CA, United States
| | - Evan G. Antzoulatos
- Center for Neuroscience, University of California, Davis, Davis, CA, United States
- Department of Neurobiology, Physiology and Behavior, University of California, Davis, Davis, CA, United States
| | - Diasynou Fioravante
- Center for Neuroscience, University of California, Davis, Davis, CA, United States
- Department of Neurobiology, Physiology and Behavior, University of California, Davis, Davis, CA, United States
- *Correspondence: Diasynou Fioravante
| |
Collapse
|
14
|
Hwang KD, Kim SJ, Lee YS. Cerebellar Circuits for Classical Fear Conditioning. Front Cell Neurosci 2022; 16:836948. [PMID: 35431810 PMCID: PMC9005982 DOI: 10.3389/fncel.2022.836948] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 03/02/2022] [Indexed: 11/17/2022] Open
Abstract
Accumulating evidence indicates that the cerebellum is critically involved in modulating non-motor behaviors, including cognition and emotional processing. Both imaging and lesion studies strongly suggest that the cerebellum is a component of the fear memory network. Given the well-established role of the cerebellum in adaptive prediction of movement and cognition, the cerebellum is likely to be engaged in the prediction of learned threats. The cerebellum is activated by fear learning, and fear learning induces changes at multiple synaptic sites in the cerebellum. Furthermore, recent technological advances have enabled the investigation of causal relationships between intra- and extra-cerebellar circuits and fear-related behaviors such as freezing. Here, we review the literature on the mechanisms underlying the modulation of cerebellar circuits in a mammalian brain by fear conditioning at the cellular and synaptic levels to elucidate the contributions of distinct cerebellar structures to fear learning and memory. This knowledge may facilitate a deeper understanding and development of more effective treatment strategies for fear-related affective disorders including post-traumatic stress or anxiety related disorders.
Collapse
Affiliation(s)
- Kyoung-Doo Hwang
- Department of Physiology, Seoul National University College of Medicine, Seoul, South Korea
- Department of Biomedical Science, Seoul National University College of Medicine, Seoul, South Korea
| | - Sang Jeong Kim
- Department of Physiology, Seoul National University College of Medicine, Seoul, South Korea
- Department of Biomedical Science, Seoul National University College of Medicine, Seoul, South Korea
- Neuroscience Research Institute, Seoul National University College of Medicine, Seoul, South Korea
- Wide River Institute of Immunology, Seoul National University, Hongcheon, South Korea
| | - Yong-Seok Lee
- Department of Physiology, Seoul National University College of Medicine, Seoul, South Korea
- Department of Biomedical Science, Seoul National University College of Medicine, Seoul, South Korea
- Neuroscience Research Institute, Seoul National University College of Medicine, Seoul, South Korea
- Wide River Institute of Immunology, Seoul National University, Hongcheon, South Korea
- *Correspondence: Yong-Seok Lee
| |
Collapse
|
15
|
Gawande DY, Kumar S Narasimhan K, Bhatt JM, Pavuluri R, Kesherwani V, Suryavanshi PS, Shelkar GP, Dravid SM. Glutamate delta 1 receptor regulates autophagy mechanisms and affects excitatory synapse maturation in the somatosensory cortex. Pharmacol Res 2022; 178:106144. [PMID: 35304260 PMCID: PMC9090310 DOI: 10.1016/j.phrs.2022.106144] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 02/01/2022] [Accepted: 02/22/2022] [Indexed: 10/18/2022]
Abstract
The glutamate delta family of receptors is composed of GluD1 and GluD2 and serve as synaptic organizers. We have previously demonstrated several autism-like molecular and behavioral phenotypes including an increase in dendritic spines in GluD1 knockout mice. Based on previous reports we evaluated whether disruption of autophagy mechanisms may account for these phenotypes. Mouse model with conditional deletion of GluD1 from excitatory neurons in the corticolimbic regions was utilized. GluD1 loss led to overactive Akt-mTOR pathway, higher p62 and a lower LC3-II/LC3-I ratio in the somatosensory cortex suggesting reduced autophagy. Excitatory elements were increased in number but had immature phenotype based on puncta size, lower AMPA subunit GluA1 expression and impaired development switch from predominantly GluN2B to mixed GluN2A/GluN2B subunit expression. Overactive Akt-mTOR signaling and impaired autophagy was also observed in dorsal striatum upon conditional ablation of GluD1 and in the prefrontal cortex and hippocampus in constitutive knockout. Finally, cognitive deficits in novel object recognition test and fear conditioning were observed in mice with conditional ablation of GluD1 from the corticolimbic regions. Together, these results demonstrate a novel function of GluD1 in the regulation of autophagy pathway which may underlie autism phenotypes and is relevant to the genetic association of GluD1 coding, GRID1 gene with autism and other developmental disorders.
Collapse
Affiliation(s)
- Dinesh Y Gawande
- Department of Pharmacology and Neuroscience, Creighton University, 2500 California Plaza, Omaha, NE, USA.
| | - Kishore Kumar S Narasimhan
- Department of Pharmacology and Neuroscience, Creighton University, 2500 California Plaza, Omaha, NE, USA
| | - Jay M Bhatt
- Department of Pharmacology and Neuroscience, Creighton University, 2500 California Plaza, Omaha, NE, USA
| | - Ratnamala Pavuluri
- Department of Pharmacology and Neuroscience, Creighton University, 2500 California Plaza, Omaha, NE, USA
| | - Varun Kesherwani
- Department of Pharmacology and Neuroscience, Creighton University, 2500 California Plaza, Omaha, NE, USA
| | - Pratyush S Suryavanshi
- Department of Pharmacology and Neuroscience, Creighton University, 2500 California Plaza, Omaha, NE, USA
| | - Gajanan P Shelkar
- Department of Pharmacology and Neuroscience, Creighton University, 2500 California Plaza, Omaha, NE, USA
| | - Shashank M Dravid
- Department of Pharmacology and Neuroscience, Creighton University, 2500 California Plaza, Omaha, NE, USA.
| |
Collapse
|
16
|
Cerebellum and Emotion Memory. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1378:53-73. [DOI: 10.1007/978-3-030-99550-8_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
17
|
Cerebellin-2 regulates a serotonergic dorsal raphe circuit that controls compulsive behaviors. Mol Psychiatry 2021; 26:7509-7521. [PMID: 34158618 PMCID: PMC8692491 DOI: 10.1038/s41380-021-01187-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 06/01/2021] [Indexed: 12/11/2022]
Abstract
Cerebellin-1 (Cbln1) and cerebellin-2 (Cbln2) are secreted glycoproteins that are expressed in distinct subsets of neurons throughout the brain. Cbln1 and Cbln2 simultaneously bind to presynaptic neurexins and postsynaptic GluD1 and GluD2, thereby forming trans-synaptic adhesion complexes. Genetic associations link cerebellins, neurexins and GluD's to neuropsychiatric disorders involving compulsive behaviors, such as Tourette syndrome, attention-deficit hyperactivity disorder (ADHD), and obsessive-compulsive disorder (OCD). Extensive evidence implicates dysfunction of serotonergic signaling in these neuropsychiatric disorders. Here, we report that constitutive Cbln2 KO mice, but not Cbln1 KO mice, display robust compulsive behaviors, including stereotypic pattern running, marble burying, explosive jumping, and excessive nest building, and exhibit decreased brain serotonin levels. Strikingly, treatment of Cbln2 KO mice with the serotonin precursor 5-hydroxytryptophan or the serotonin reuptake-inhibitor fluoxetine alleviated compulsive behaviors. Conditional deletion of Cbln2 both from dorsal raphe neurons and from presynaptic neurons synapsing onto dorsal raphe neurons reproduced the compulsive behaviors of Cbln2 KO mice. Finally, injection of recombinant Cbln2 protein into the dorsal raphe of Cbln2 KO mice largely reversed their compulsive behaviors. Taken together, our results show that Cbln2 controls compulsive behaviors by regulating serotonergic circuits in the dorsal raphe.
Collapse
|
18
|
Gandhi PJ, Gawande DY, Shelkar GP, Gakare SG, Kiritoshi T, Ji G, Misra B, Pavuluri R, Liu J, Neugebauer V, Dravid SM. Dysfunction of Glutamate Delta-1 Receptor-Cerebellin 1 Trans-Synaptic Signaling in the Central Amygdala in Chronic Pain. Cells 2021; 10:2644. [PMID: 34685624 PMCID: PMC8534524 DOI: 10.3390/cells10102644] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 09/29/2021] [Accepted: 09/30/2021] [Indexed: 01/02/2023] Open
Abstract
Chronic pain is a debilitating condition involving neuronal dysfunction, but the synaptic mechanisms underlying the persistence of pain are still poorly understood. We found that the synaptic organizer glutamate delta 1 receptor (GluD1) is expressed postsynaptically at parabrachio-central laterocapsular amygdala (PB-CeLC) glutamatergic synapses at axo-somatic and punctate locations on protein kinase C δ -positive (PKCδ+) neurons. Deletion of GluD1 impairs excitatory neurotransmission at the PB-CeLC synapses. In inflammatory and neuropathic pain models, GluD1 and its partner cerebellin 1 (Cbln1) are downregulated while AMPA receptor is upregulated. A single infusion of recombinant Cbln1 into the central amygdala led to sustained mitigation of behavioral pain parameters and normalized hyperexcitability of central amygdala neurons. Cbln2 was ineffective under these conditions and the effect of Cbln1 was antagonized by GluD1 ligand D-serine. The behavioral effect of Cbln1 was GluD1-dependent and showed lateralization to the right central amygdala. Selective ablation of GluD1 from the central amygdala or injection of Cbln1 into the central amygdala in normal animals led to changes in averse and fear-learning behaviors. Thus, GluD1-Cbln1 signaling in the central amygdala is a teaching signal for aversive behavior but its sustained dysregulation underlies persistence of pain. Significance statement: Chronic pain is a debilitating condition which involves synaptic dysfunction, but the underlying mechanisms are not fully understood. Our studies identify a novel mechanism involving structural synaptic changes in the amygdala caused by impaired GluD1-Cbln1 signaling in inflammatory and neuropathic pain behaviors. We also identify a novel means to mitigate pain in these conditions using protein therapeutics.
Collapse
Affiliation(s)
- Pauravi J. Gandhi
- Department of Pharmacology and Neuroscience, Creighton University School of Medicine, Omaha, NE 68178, USA; (P.J.G.); (D.Y.G.); (G.P.S.); (S.G.G.); (B.M.); (R.P.); (J.L.)
| | - Dinesh Y. Gawande
- Department of Pharmacology and Neuroscience, Creighton University School of Medicine, Omaha, NE 68178, USA; (P.J.G.); (D.Y.G.); (G.P.S.); (S.G.G.); (B.M.); (R.P.); (J.L.)
| | - Gajanan P. Shelkar
- Department of Pharmacology and Neuroscience, Creighton University School of Medicine, Omaha, NE 68178, USA; (P.J.G.); (D.Y.G.); (G.P.S.); (S.G.G.); (B.M.); (R.P.); (J.L.)
| | - Sukanya G. Gakare
- Department of Pharmacology and Neuroscience, Creighton University School of Medicine, Omaha, NE 68178, USA; (P.J.G.); (D.Y.G.); (G.P.S.); (S.G.G.); (B.M.); (R.P.); (J.L.)
| | - Takaki Kiritoshi
- Department of Pharmacology and Neuroscience, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; (T.K.); (G.J.); (V.N.)
| | - Guangchen Ji
- Department of Pharmacology and Neuroscience, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; (T.K.); (G.J.); (V.N.)
- Center of Excellence for Translational Neuroscience and Therapeutics, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Bishal Misra
- Department of Pharmacology and Neuroscience, Creighton University School of Medicine, Omaha, NE 68178, USA; (P.J.G.); (D.Y.G.); (G.P.S.); (S.G.G.); (B.M.); (R.P.); (J.L.)
| | - Ratnamala Pavuluri
- Department of Pharmacology and Neuroscience, Creighton University School of Medicine, Omaha, NE 68178, USA; (P.J.G.); (D.Y.G.); (G.P.S.); (S.G.G.); (B.M.); (R.P.); (J.L.)
| | - Jinxu Liu
- Department of Pharmacology and Neuroscience, Creighton University School of Medicine, Omaha, NE 68178, USA; (P.J.G.); (D.Y.G.); (G.P.S.); (S.G.G.); (B.M.); (R.P.); (J.L.)
| | - Volker Neugebauer
- Department of Pharmacology and Neuroscience, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; (T.K.); (G.J.); (V.N.)
- Center of Excellence for Translational Neuroscience and Therapeutics, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- Garrison Institute on Aging, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Shashank M. Dravid
- Department of Pharmacology and Neuroscience, Creighton University School of Medicine, Omaha, NE 68178, USA; (P.J.G.); (D.Y.G.); (G.P.S.); (S.G.G.); (B.M.); (R.P.); (J.L.)
| |
Collapse
|
19
|
Wang F, Wang Q, Liu B, Mei L, Ma S, Wang S, Wang R, Zhang Y, Niu C, Xiong Z, Zheng Y, Zhang Z, Shi J, Song X. The long noncoding RNA Synage regulates synapse stability and neuronal function in the cerebellum. Cell Death Differ 2021; 28:2634-2650. [PMID: 33762741 PMCID: PMC8408218 DOI: 10.1038/s41418-021-00774-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 03/07/2021] [Accepted: 03/10/2021] [Indexed: 02/01/2023] Open
Abstract
The brain is known to express many long noncoding RNAs (lncRNAs); however, whether and how these lncRNAs function in modulating synaptic stability remains unclear. Here, we report a cerebellum highly expressed lncRNA, Synage, regulating synaptic stability via at least two mechanisms. One is through the function of Synage as a sponge for the microRNA miR-325-3p, to regulate expression of the known cerebellar synapse organizer Cbln1. The other function is to serve as a scaffold for organizing the assembly of the LRP1-HSP90AA1-PSD-95 complex in PF-PC synapses. Although somewhat divergent in its mature mRNA sequence, the locus encoding Synage is positioned adjacent to the Cbln1 loci in mouse, rhesus macaque, and human, and Synage is highly expressed in the cerebella of all three species. Synage deletion causes a full-spectrum cerebellar ablation phenotype that proceeds from cerebellar atrophy, through neuron loss, on to synapse density reduction, synaptic vesicle loss, and finally to a reduction in synaptic activity during cerebellar development; these deficits are accompanied by motor dysfunction in adult mice, which can be rescued by AAV-mediated Synage overexpression from birth. Thus, our study demonstrates roles for the lncRNA Synage in regulating synaptic stability and function during cerebellar development.
Collapse
Affiliation(s)
- Fei Wang
- grid.59053.3a0000000121679639Hefei National Laboratory for Physical Sciences at the Microscale, CAS Key Laboratory of Brain Function and Disease, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui China
| | - Qianqian Wang
- grid.59053.3a0000000121679639Hefei National Laboratory for Physical Sciences at the Microscale, CAS Key Laboratory of Brain Function and Disease, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui China
| | - Baowei Liu
- grid.59053.3a0000000121679639Hefei National Laboratory for Physical Sciences at the Microscale, CAS Key Laboratory of Brain Function and Disease, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui China
| | - Lisheng Mei
- grid.59053.3a0000000121679639Hefei National Laboratory for Physical Sciences at the Microscale, CAS Key Laboratory of Brain Function and Disease, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui China
| | - Sisi Ma
- grid.506261.60000 0001 0706 7839National Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, CAMS and PUMC, Beijing, China
| | - Shujuan Wang
- grid.419611.a0000 0004 0457 9072State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
| | - Ruoyu Wang
- grid.59053.3a0000000121679639Hefei National Laboratory for Physical Sciences at the Microscale, CAS Key Laboratory of Brain Function and Disease, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui China ,grid.240145.60000 0001 2291 4776Graduate School of Biomedical Sciences, University of Texas MD Anderson Cancer Center and UTHealth, Houston, TX USA
| | - Yan Zhang
- grid.59053.3a0000000121679639Stroke Center & Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui China
| | - Chaoshi Niu
- grid.59053.3a0000000121679639Department of Neurosurgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui China
| | - Zhiqi Xiong
- grid.9227.e0000000119573309Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Yong Zheng
- grid.419611.a0000 0004 0457 9072State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
| | - Zhi Zhang
- grid.59053.3a0000000121679639Hefei National Laboratory for Physical Sciences at the Microscale, CAS Key Laboratory of Brain Function and Disease, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui China
| | - Juan Shi
- grid.506261.60000 0001 0706 7839National Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, CAMS and PUMC, Beijing, China
| | - Xiaoyuan Song
- grid.59053.3a0000000121679639MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, Hefei National Laboratory for Physical Sciences at the Microscale, CAS Key Laboratory of Brain Function and Disease, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| |
Collapse
|
20
|
Larsen K. The porcine cerebellin gene family. Gene 2021; 799:145852. [PMID: 34274480 DOI: 10.1016/j.gene.2021.145852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 07/13/2021] [Indexed: 11/18/2022]
Abstract
Cerebellins (CBLN1-4), together with C1qTNF proteins, belong to the CBLN subfamily of C1q proteins. Cerebellin-1 (CBLN1) is active in synapse formation and functions at the parallel fiber-Purkinje cell synapses. Cerebellins form tripartite complexes with neurexins and the glutamate-receptor-related proteins GluD1 and GluD2, playing a role as trans-synaptic cell-adhesion molecules that critically contribute to both synapse formation and functioning and brain development. In this study, I present a molecular characterization of the four porcine CBLN genes. Experimental data and in silico analyses collectively describes the gene structure, chromosomal localization, and expression of CBLN1-4. Two cDNAs encoding the cerebellins CBLN1 and CBLN3 were RT-PCR cloned and sequenced. The nucleotide sequence of the CBLN1 clone contains an open reading frame of 582 nucleotides and encodes a protein of 193 amino acids. The deduced amino acid of the porcine CBLN1 protein was 99% identical to both mouse CBLN1 and to human CBLN1. The deduced CBLN1 protein contains a putative signal sequence of 21 residues, two conserved cysteine residues, and C1q domain. The nucleotide sequence of the CBLN3 cDNA clone comprises an open reading frame of 618 nucleotides and encodes a protein of 205 amino acids. The deduced amino acid sequence of the porcine CBLN3 protein was 88% identical to mouse CBLN3 and 94% identical to human CBLN3. The amino terminal ends of both the CBLN1 and CBLN3 proteins contain three possible N-linked glycosylation sites. The genomic organization of both porcine CBLN1 and CBLN3 is very similar to those of their human counterparts. The expression analyses demonstrated that CBLN1 and CBLN3 transcripts are predominantly expressed in the cerebellum. The sequences of the porcine precerebellin genes and cDNAs were submitted to DDBJ/EMBL/GenBank under the following accession numbers: CBLN1 gene (GenBank ID: FJ621565), CBLN1 cDNA (GenBank ID: EF577504), CBLN3 gene (GenBank ID: FJ621566), CBLN3 cDNA (GenBank ID: EF577505) and CBLN4 cDNA (GenBank ID: FJ196070).
Collapse
Affiliation(s)
- Knud Larsen
- Department of Molecular Biology and Genetics, Aarhus University, C.F. Møllers Allé 3, DK-8000 Aarhus C, Denmark.
| |
Collapse
|
21
|
Long noncoding RNA PM maintains cerebellar synaptic integrity and Cbln1 activation via Pax6/Mll1-mediated H3K4me3. PLoS Biol 2021; 19:e3001297. [PMID: 34111112 PMCID: PMC8219131 DOI: 10.1371/journal.pbio.3001297] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Revised: 06/22/2021] [Accepted: 05/24/2021] [Indexed: 01/30/2023] Open
Abstract
Recent studies have shown that long noncoding RNAs (lncRNAs) are critical regulators in the central nervous system (CNS). However, their roles in the cerebellum are currently unclear. In this work, we identified the isoform 204 of lncRNA Gm2694 (designated as lncRNA-Promoting Methylation (lncRNA-PM)) is highly expressed in the cerebellum and derived from the antisense strand of the upstream region of Cerebellin-1 (Cbln1), a well-known critical cerebellar synaptic organizer. LncRNA-PM exhibits similar spatiotemporal expression pattern as Cbln1 in the postnatal mouse cerebellum and activates the transcription of Cbln1 through Pax6/Mll1-mediated H3K4me3. In mouse cerebellum, lncRNA-PM, Pax6/Mll1, and H3K4me3 are all associated with the regulatory regions of Cbln1. Knockdown of lncRNA-PM in cerebellum causes deficiencies in Cbln1 expression, cerebellar synaptic integrity, and motor function. Together, our work reveals an lncRNA-mediated transcriptional activation of Cbln1 through Pax6-Mll1-H3K4me3 and provides novel insights of the essential roles of lncRNA in the cerebellum. The long non-coding RNA lncRNA-PM activates transcription of the cerebellar synaptic organizer Cbln1 by promoting Pax6-Mll1-mediated H3K4me3 methylation, thereby helping to maintain cerebellar synaptic integrity and motor function.
Collapse
|
22
|
Andrews PC, Dravid SM. An emerging map of glutamate delta 1 receptors in the forebrain. Neuropharmacology 2021; 192:108587. [PMID: 33992669 DOI: 10.1016/j.neuropharm.2021.108587] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 04/15/2021] [Accepted: 04/20/2021] [Indexed: 11/19/2022]
Abstract
Glutamate delta 1 (GluD1) and glutamate delta 2 (GluD2) form the delta family of ionotropic glutamate receptors; these proteins plays widespread roles in synaptic architecture, motor behavior, and cognitive function. Though the role of GluD2 at cerebellar parallel fiber-Purkinje cell synapses is well established, attention now turns to the function of GluD receptors in the forebrain. GluD1 regulates synaptic assembly and modulation in multiple higher brain regions, acting as a postsynaptic cell adhesion molecule with effects on both excitatory and inhibitory transmission. Furthermore, variations and mutations in the GRID1 gene, which codes for GluD1, and in genes which code for proteins functionally linked to GluD1, are associated with mental disorders including autism, schizophrenia, bipolar disorder, and major depression. Cerebellin (Cbln) family proteins, the primary binding partners of delta receptors, are secreted C1q-like proteins which also bind presynaptic neurexins (NRXNs), forming a tripartite synaptic bridge. Published research explores this bridge's function in regions including the striatum, hippocampus, cortex, and cerebellum. In this review, we summarize region- and circuit-specific functions and expression patterns for GluD1 and its related proteins, and their implications for behavior and disease.
Collapse
Affiliation(s)
- Patrick C Andrews
- Department of Pharmacology and Neuroscience, Creighton University, 2500 California Plaza, Omaha, NE, USA
| | - Shashank M Dravid
- Department of Pharmacology and Neuroscience, Creighton University, 2500 California Plaza, Omaha, NE, USA.
| |
Collapse
|
23
|
Kim HY, Um JW, Ko J. Proper synaptic adhesion signaling in the control of neural circuit architecture and brain function. Prog Neurobiol 2021; 200:101983. [PMID: 33422662 DOI: 10.1016/j.pneurobio.2020.101983] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 11/23/2020] [Accepted: 12/22/2020] [Indexed: 12/17/2022]
Abstract
Trans-synaptic cell-adhesion molecules are critical for governing various stages of synapse development and specifying neural circuit properties via the formation of multifarious signaling pathways. Recent studies have pinpointed the putative roles of trans-synaptic cell-adhesion molecules in mediating various cognitive functions. Here, we review the literature on the roles of a diverse group of central synaptic organizers, including neurexins (Nrxns), leukocyte common antigen-related receptor protein tyrosine phosphatases (LAR-RPTPs), and their associated binding proteins, in regulating properties of specific type of synapses and neural circuits. In addition, we highlight the findings that aberrant synaptic adhesion signaling leads to alterations in the structures, transmission, and plasticity of specific synapses across diverse brain areas. These results seem to suggest that proper trans-synaptic signaling pathways by Nrxns, LAR-RPTPs, and their interacting network is likely to constitute central molecular complexes that form the basis for cognitive functions, and that these complexes are heterogeneously and complexly disrupted in many neuropsychiatric and neurodevelopmental disorders.
Collapse
Affiliation(s)
- Hee Young Kim
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, 42988, South Korea
| | - Ji Won Um
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, 42988, South Korea; Core Protein Resources Center, DGIST, Daegu, 42988, South Korea.
| | - Jaewon Ko
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, 42988, South Korea.
| |
Collapse
|
24
|
Wang Y, Zhang S, Yang H, Zhang X, He S, Wang J, Li J. Altered cerebellum functional network on newly diagnosed drug-naïve Parkinson's disease patients with anxiety. Transl Neurosci 2021; 12:415-424. [PMID: 34760297 PMCID: PMC8556613 DOI: 10.1515/tnsci-2020-0192] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 09/22/2021] [Accepted: 10/08/2021] [Indexed: 01/08/2023] Open
Abstract
Introduction Damage to the cerebellar functional network may underlie anxiety symptoms in patients with Parkinson’s disease (PD). Herein we investigated the regional homogeneity (ReHo) and functional connectivity (FC) patterns of cerebellar and clinical correlates in PD patients with anxiety and explored their clinical significance. Methods We enrolled 50 newly diagnosed drug-naïve PD patients and 30 normal controls (NCs). Twenty-six PD patients with anxiety symptoms (PD-A) and 24 PD patients without anxiety symptoms (PD-NA) were sorted into groups based on the Hamilton Anxiety Scale (HAMA). All included participants underwent rest-state functional magnetic resonance imaging (rs-fMRI) scanning. Cerebellar FC based on the seed-based method was used to investigate regional and whole brain function in PD-A, PD-NA, and NCs, and the relationship between the abnormal brain function and anxiety symptoms in PD patients was also detected. Results Compared with the PD-NA group and the NCs, the ReHo value of the PD-A group was significantly decreased in the left medial frontal gyrus and increased in the left cerebellum. Further, left-cerebellum-based FC patterns were used to detect the decreased FC in the right cerebellum, while FC was increased in the right caudate nucleus, and the right anterior cingulate cortex (ACC) in the PD-A group was compared with that in the PD-NA group. Further, the altered FC between the left cerebellum and the right cerebellum was significantly associated with anxiety symptoms in the PD-A group. Conclusion The present study found abnormal regional cerebellum function as well as disruptions in the connectivity network within the cerebellum, caudate, and ACC in patients with PD-A. In addition, the FC between the left cerebellum and the right cerebellum was associated with anxiety symptoms in patients with PD. The present study indicated that cerebellar functional damage may be associated with anxiety symptoms in PD patients.
Collapse
Affiliation(s)
- Yirong Wang
- Department of Neurology, Chengdu Second People's Hospital, Chengdu, Sichuan Province, People's Republic of China.,Department of Neurology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan Province, People's Republic of China
| | - Shushan Zhang
- Department of Neurology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan Province, People's Republic of China
| | - Haodi Yang
- Department of Neurology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan Province, People's Republic of China
| | - Xin Zhang
- Department of Neurology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan Province, People's Republic of China
| | - Shijia He
- Department of Neurology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan Province, People's Republic of China
| | - Jian Wang
- Department of Neurology, Chengdu Second People's Hospital, Chengdu, Sichuan Province, People's Republic of China
| | - Jian Li
- North Sichuan Medical College, No. 234, Fujiang Street, Shunqing District, Nanchong, Sichuan Province, People's Republic of China
| |
Collapse
|
25
|
Lee YJ, Guell X, Hubbard NA, Siless V, Frosch IR, Goncalves M, Lo N, Nair A, Ghosh SS, Hofmann SG, Auerbach RP, Pizzagalli DA, Yendiki A, Gabrieli JDE, Whitfield-Gabrieli S, Anteraper SA. Functional Alterations in Cerebellar Functional Connectivity in Anxiety Disorders. THE CEREBELLUM 2020; 20:392-401. [PMID: 33210245 PMCID: PMC8213597 DOI: 10.1007/s12311-020-01213-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Accepted: 11/08/2020] [Indexed: 01/24/2023]
Abstract
Adolescents with anxiety disorders exhibit excessive emotional and somatic arousal. Neuroimaging studies have shown abnormal cerebral cortical activation and connectivity in this patient population. The specific role of cerebellar output circuitry, specifically the dentate nuclei (DN), in adolescent anxiety disorders remains largely unexplored. Resting-state functional connectivity analyses have parcellated the DN, the major output nuclei of the cerebellum, into three functional territories (FTs) that include default-mode, salience-motor, and visual networks. The objective of this study was to understand whether FTs of the DN are implicated in adolescent anxiety disorders. Forty-one adolescents (mean age 15.19 ± 0.82, 26 females) with one or more anxiety disorders and 55 age- and gender-matched healthy controls completed resting-state fMRI scans and a self-report survey on anxiety symptoms. Seed-to-voxel functional connectivity analyses were performed using the FTs from DN parcellation. Brain connectivity metrics were then correlated with State-Trait Anxiety Inventory (STAI) measures within each group. Adolescents with an anxiety disorder showed significant hyperconnectivity between salience-motor DN FT and cerebral cortical salience-motor regions compared to controls. Salience-motor FT connectivity with cerebral cortical sensorimotor regions was significantly correlated with STAI-trait scores in HC (R2 = 0.41). Here, we report DN functional connectivity differences in adolescents diagnosed with anxiety, as well as in HC with variable degrees of anxiety traits. These observations highlight the relevance of DN as a potential clinical and sub-clinical marker of anxiety.
Collapse
Affiliation(s)
- Yoon Ji Lee
- Department of Psychology, ISEC 672D, Northeastern University, Boston, MA, 02115, USA
| | | | - Nicholas A Hubbard
- University of Nebraska-Lincoln, Lincoln, NE, USA.,Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Viviana Siless
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Boston, MA, USA
| | | | | | - Nicole Lo
- Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Atira Nair
- Department of Psychology, ISEC 672D, Northeastern University, Boston, MA, 02115, USA
| | - Satrajit S Ghosh
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA
| | | | | | | | - Anastasia Yendiki
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA
| | | | - Susan Whitfield-Gabrieli
- Department of Psychology, ISEC 672D, Northeastern University, Boston, MA, 02115, USA.,Massachusetts Institute of Technology, Cambridge, MA, USA
| | | |
Collapse
|
26
|
Hoover AH, Pavuluri R, Shelkar GP, Dravid SM, Smith Y, Villalba RM. Ultrastructural localization of glutamate delta 1 (GluD1) receptor immunoreactivity in the mouse and monkey striatum. J Comp Neurol 2020; 529:1703-1718. [PMID: 33084025 DOI: 10.1002/cne.25051] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 10/04/2020] [Accepted: 10/05/2020] [Indexed: 12/26/2022]
Abstract
The glutamate receptor delta 1 (GluD1) is strongly expressed in the striatum. Knockout of GluD1 expression in striatal neurons elicits cognitive deficits and disrupts the thalamostriatal system in mice. To understand the potential role of GluD1 in the primate striatum, we compared the cellular and subcellular localization of striatal GluD1 immunoreactivity (GluD1-IR) in mice and monkeys. In both species, striatal GluD1-IR displayed a patchy pattern of distribution in register with the striosome/matrix compartmentation, but in an opposite fashion. While GluD1 was more heavily expressed in the striosomes than the matrix in the monkey caudate nucleus, the opposite was found in the mouse striatum. At the electron microscopic level, GluD1-IR was preferentially expressed in dendritic shafts (47.9 ± 1.2%), followed by glia (37.7 ± 2.5%), and dendritic spines (14.3 ± 2.6%) in the matrix of the mouse striatum. This pattern was not statistically different from the labeling in the striosome and matrix compartments of the monkey caudate nucleus, with the exception of a small amount of GluD1-positive unmyelinated axons and axon terminals in the primate striatum. Immunogold staining revealed synaptic and perisynaptic GluD1 labeling at putative axo-dendritic and axo-spinous glutamatergic synapses, and intracellular labeling on the surface of mitochondria. Confocal microscopy showed that GluD1 is preferentially colocalized with thalamic over cortical terminals in both the striosome and matrix compartments. These data provide the anatomical substrate for a deeper understanding of GluD1 regulation of striatal glutamatergic synapses, but also suggest possible extrasynaptic, glial, and mitochondrial GluD1 functions.
Collapse
Affiliation(s)
- Andrew H Hoover
- Yerkes National Primate Research Center, Atlanta, Georgia, USA.,UDALL Center of Excellence for Parkinson's Disease, Atlanta, Georgia, USA
| | - Ratnamala Pavuluri
- Department of Pharmacology and Neuroscience, Creighton University School of Medicine, Omaha, Nebraska, USA
| | - Gajanan P Shelkar
- Department of Pharmacology and Neuroscience, Creighton University School of Medicine, Omaha, Nebraska, USA
| | - Shashank M Dravid
- Department of Pharmacology and Neuroscience, Creighton University School of Medicine, Omaha, Nebraska, USA
| | - Yoland Smith
- Yerkes National Primate Research Center, Atlanta, Georgia, USA.,UDALL Center of Excellence for Parkinson's Disease, Atlanta, Georgia, USA.,Department of Neurology, Emory University, Atlanta, Georgia, USA
| | - Rosa M Villalba
- Yerkes National Primate Research Center, Atlanta, Georgia, USA.,UDALL Center of Excellence for Parkinson's Disease, Atlanta, Georgia, USA
| |
Collapse
|
27
|
MeCP2 Levels Regulate the 3D Structure of Heterochromatic Foci in Mouse Neurons. J Neurosci 2020; 40:8746-8766. [PMID: 33046553 DOI: 10.1523/jneurosci.1281-19.2020] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2019] [Revised: 08/25/2020] [Accepted: 09/17/2020] [Indexed: 02/02/2023] Open
Abstract
Methyl-CpG binding protein 2 (MeCP2) is a nuclear protein critical for normal brain function, and both depletion and overexpression of MeCP2 lead to severe neurodevelopmental disease, Rett syndrome (RTT) and MECP2 multiplication disorder, respectively. However, the molecular mechanism by which abnormal MeCP2 dosage causes neuronal dysfunction remains unclear. As MeCP2 expression is nearly equivalent to that of core histones and because it binds DNA throughout the genome, one possible function of MeCP2 is to regulate the 3D structure of chromatin. Here, to examine whether and how MeCP2 levels impact chromatin structure, we used high-resolution confocal and electron microscopy and examined heterochromatic foci of neurons in mice. Using models of RTT and MECP2 triplication syndrome, we found that the heterochromatin structure was significantly affected by the alteration in MeCP2 levels. Analysis of mice expressing either MeCP2-R270X or MeCP2-G273X, which have nonsense mutations in the upstream and downstream regions of the AT-hook 2 domain, respectively, showed that the magnitude of heterochromatin changes was tightly correlated with the phenotypic severity. Postnatal alteration in MeCP2 levels also induced significant changes in the heterochromatin structure, which underscored importance of correct MeCP2 dosage in mature neurons. Finally, functional analysis of MeCP2-overexpressing mice showed that the behavioral and transcriptomic alterations in these mice correlated significantly with the MeCP2 levels and occurred in parallel with the heterochromatin changes. Taken together, our findings demonstrate the essential role of MeCP2 in regulating the 3D structure of neuronal chromatin, which may serve as a potential mechanism that drives pathogenesis of MeCP2-related disorders.SIGNIFICANCE STATEMENT Neuronal function is critically dependent on methyl-CpG binding protein 2 (MeCP2), a nuclear protein abundantly expressed in neurons. The importance of MeCP2 is underscored by the severe childhood neurologic disorders, Rett syndrome (RTT) and MECP2 multiplication disorders, which are caused by depletion and overabundance of MeCP2, respectively. To clarify the molecular function of MeCP2 and to understand the pathogenesis of MECP2-related disorders, we performed detailed structural analyses of neuronal nuclei by using mouse models and high-resolution microscopy. We show that the level of MeCP2 critically regulates 3D structure of heterochromatic foci, and this is mediated in part by the AT-hook 2 domain of MeCP2. Our results demonstrate that one primary function of MeCP2 is to regulate chromatin structure.
Collapse
|
28
|
Zhang L, Song NN, Zhang Q, Mei WY, He CH, Ma P, Huang Y, Chen JY, Mao B, Lang B, Ding YQ. Satb2 is required for the regionalization of retrosplenial cortex. Cell Death Differ 2020; 27:1604-1617. [PMID: 31666685 PMCID: PMC7206047 DOI: 10.1038/s41418-019-0443-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2019] [Revised: 10/10/2019] [Accepted: 10/11/2019] [Indexed: 02/08/2023] Open
Abstract
The retrosplenial cortex (Rsp) is a transitional cortex located between the neocortex and archicortex, but the molecular mechanism specifying Rsp from the archicortex remains elusive. We here report that the transcription factor Satb2 is required for specifying Rsp identity during its morphogenesis. In Satb2 CKO mice, the boundary between the Rsp and archicortex [i.e., subiculum (SubC)] disappears as early as E17.5, and Rsp efferent projection is aberrant. Rsp-specific genes are lost, whereas SubC-specific genes are ectopically expressed in Rsp of Satb2 CKO mice. Furthermore, cell-autonomous role of Satb2 in maintaining Rsp neuron identity is revealed by inactivation of Satb2 in Rsp neurons. Finally, Satb2 represses the transcription of Nr4a2. The misexpression of Nr4a2 together with Ctip2 induces expression of SubC-specific genes in wild-type Rsp, and simultaneous knockdown of these two genes in Rsp Satb2-mutant cells prevents their fate transition to SubC identity. Thus, Satb2 serves as a determinant gene in the Rsp regionalization by repressing Nr4a2 and Ctip2 during cortical development.
Collapse
Affiliation(s)
- Lei Zhang
- Key Laboratory of Arrhythmias, Ministry of Education of China, East Hospital, and Department of Anatomy and Neurobiology, Collaborative Innovation Center for Brain Science, Tongji University School of Medicine, Shanghai, 200092, China.
| | - Ning-Ning Song
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Qiong Zhang
- Key Laboratory of Arrhythmias, Ministry of Education of China, East Hospital, and Department of Anatomy and Neurobiology, Collaborative Innovation Center for Brain Science, Tongji University School of Medicine, Shanghai, 200092, China
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Wan-Ying Mei
- Key Laboratory of Arrhythmias, Ministry of Education of China, East Hospital, and Department of Anatomy and Neurobiology, Collaborative Innovation Center for Brain Science, Tongji University School of Medicine, Shanghai, 200092, China
| | - Chun-Hui He
- Key Laboratory of Arrhythmias, Ministry of Education of China, East Hospital, and Department of Anatomy and Neurobiology, Collaborative Innovation Center for Brain Science, Tongji University School of Medicine, Shanghai, 200092, China
| | - Pengcheng Ma
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China
- Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, 650223, China
| | - Ying Huang
- Key Laboratory of Arrhythmias, Ministry of Education of China, East Hospital, and Department of Anatomy and Neurobiology, Collaborative Innovation Center for Brain Science, Tongji University School of Medicine, Shanghai, 200092, China
| | - Jia-Yin Chen
- Key Laboratory of Arrhythmias, Ministry of Education of China, East Hospital, and Department of Anatomy and Neurobiology, Collaborative Innovation Center for Brain Science, Tongji University School of Medicine, Shanghai, 200092, China
| | - Bingyu Mao
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China
- Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, 650223, China
| | - Bing Lang
- Key Laboratory of Arrhythmias, Ministry of Education of China, East Hospital, and Department of Anatomy and Neurobiology, Collaborative Innovation Center for Brain Science, Tongji University School of Medicine, Shanghai, 200092, China
- Mental Health Institute of the Second Xiangya Hospital, National Clinical Research Center on Mental Disorders, National Technology Institute on Mental Disorders, Key Laboratory of Psychiatry and Mental Health of Hunan Province, Central South University, Changsha, 410011, Hunan, China
| | - Yu-Qiang Ding
- Key Laboratory of Arrhythmias, Ministry of Education of China, East Hospital, and Department of Anatomy and Neurobiology, Collaborative Innovation Center for Brain Science, Tongji University School of Medicine, Shanghai, 200092, China.
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, 200032, China.
- Department of Laboratory Animal Science, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
29
|
Nakamoto C, Kawamura M, Nakatsukasa E, Natsume R, Takao K, Watanabe M, Abe M, Takeuchi T, Sakimura K. GluD1 knockout mice with a pure C57BL/6N background show impaired fear memory, social interaction, and enhanced depressive-like behavior. PLoS One 2020; 15:e0229288. [PMID: 32078638 PMCID: PMC7032715 DOI: 10.1371/journal.pone.0229288] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Accepted: 02/03/2020] [Indexed: 01/07/2023] Open
Abstract
The GluD1 gene is associated with susceptibility for schizophrenia, autism, depression, and bipolar disorder. However, the function of GluD1 and how it is involved in these conditions remain elusive. In this study, we generated a Grid1 gene-knockout (GluD1-KO) mouse line with a pure C57BL/6N genetic background and performed several behavioral analyses. Compared to a control group, GluD1-KO mice showed no significant anxiety-related behavioral differences, evaluated using behavior in an open field, elevated plus maze, a light-dark transition test, the resident-intruder test of aggression and sensorimotor gating evaluated by the prepulse inhibition test. However, GluD1-KO mice showed (1) higher locomotor activity in the open field, (2) decreased sociability and social novelty preference in the three-chambered social interaction test, (3) impaired memory in contextual, but not cued fear conditioning tests, and (4) enhanced depressive-like behavior in a forced swim test. Pharmacological studies revealed that enhanced depressive-like behavior in GluD1-KO mice was restored by the serotonin reuptake inhibitors imipramine and fluoxetine, but not the norepinephrine transporter inhibitor desipramine. In addition, biochemical analysis revealed no significant difference in protein expression levels, such as other glutamate receptors in the synaptosome and postsynaptic densities prepared from the frontal cortex and the hippocampus. These results suggest that GluD1 plays critical roles in fear memory, sociability, and depressive-like behavior.
Collapse
Affiliation(s)
- Chihiro Nakamoto
- Department of Animal Model Development, Brain Research Institute, Niigata University, Niigata, Japan
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Danish Research Institute of Translational Neuroscience–DANDRITE, Nordic-EMBL Partnership for Molecular Medicine, Aarhus University, Aarhus, Denmark
| | - Meiko Kawamura
- Department of Animal Model Development, Brain Research Institute, Niigata University, Niigata, Japan
| | - Ena Nakatsukasa
- Department of Animal Model Development, Brain Research Institute, Niigata University, Niigata, Japan
| | - Rie Natsume
- Department of Animal Model Development, Brain Research Institute, Niigata University, Niigata, Japan
| | - Keizo Takao
- Graduate School of Innovative Life Science, University of Toyama, Toyama, Japan
- Life Science Research Center, University of Toyama, Toyama, Japan
| | - Masahiko Watanabe
- Department of Anatomy, Faculty of Medicine, Hokkaido University, Sapporo, Japan
| | - Manabu Abe
- Department of Animal Model Development, Brain Research Institute, Niigata University, Niigata, Japan
- * E-mail: (TT); (MA)
| | - Tomonori Takeuchi
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Danish Research Institute of Translational Neuroscience–DANDRITE, Nordic-EMBL Partnership for Molecular Medicine, Aarhus University, Aarhus, Denmark
- * E-mail: (TT); (MA)
| | - Kenji Sakimura
- Department of Animal Model Development, Brain Research Institute, Niigata University, Niigata, Japan
| |
Collapse
|
30
|
Liu J, Shelkar GP, Gandhi PJ, Gawande DY, Hoover A, Villalba RM, Pavuluri R, Smith Y, Dravid SM. Striatal glutamate delta-1 receptor regulates behavioral flexibility and thalamostriatal connectivity. Neurobiol Dis 2020; 137:104746. [PMID: 31945419 PMCID: PMC7204410 DOI: 10.1016/j.nbd.2020.104746] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 12/19/2019] [Accepted: 01/12/2020] [Indexed: 10/31/2022] Open
Abstract
Impaired behavioral flexibility and repetitive behavior is a common phenotype in autism and other neuropsychiatric disorders, but the underlying synaptic mechanisms are poorly understood. The trans-synaptic glutamate delta (GluD)-Cerebellin 1-Neurexin complex, critical for synapse formation/maintenance, represents a vulnerable axis for neuropsychiatric diseases. We have previously found that GluD1 deletion results in reversal learning deficit and repetitive behavior. In this study, we show that selective ablation of GluD1 from the dorsal striatum impairs behavioral flexibility in a water T-maze task. We further found that striatal GluD1 is preferentially found in dendritic shafts, and more frequently associated with thalamic than cortical glutamatergic terminals suggesting localization to projections from the thalamic parafascicular nucleus (Pf). Conditional deletion of GluD1 from the striatum led to a selective loss of thalamic, but not cortical, terminals, and reduced glutamatergic neurotransmission. Optogenetic studies demonstrated functional changes at thalamostriatal synapses from the Pf, but no effect on the corticostriatal system, upon ablation of GluD1 in the dorsal striatum. These studies suggest a novel molecular mechanism by which genetic variations associated with neuropsychiatric disorders may impair behavioral flexibility, and reveal a unique principle by which GluD1 subunit regulates forebrain circuits.
Collapse
Affiliation(s)
- Jinxu Liu
- Department of Pharmacology and Neuroscience, Creighton University School of Medicine, Omaha, NE 68178, USA
| | - Gajanan P Shelkar
- Department of Pharmacology and Neuroscience, Creighton University School of Medicine, Omaha, NE 68178, USA
| | - Pauravi J Gandhi
- Department of Pharmacology and Neuroscience, Creighton University School of Medicine, Omaha, NE 68178, USA
| | - Dinesh Y Gawande
- Department of Pharmacology and Neuroscience, Creighton University School of Medicine, Omaha, NE 68178, USA
| | - Andrew Hoover
- Yerkes National Primate Research Center, Atlanta, GA 30329, USA; UDALL Center of Excellence for Parkinson's Disease, Atlanta, GA 30329, USA
| | - Rosa M Villalba
- Yerkes National Primate Research Center, Atlanta, GA 30329, USA; UDALL Center of Excellence for Parkinson's Disease, Atlanta, GA 30329, USA
| | - Ratnamala Pavuluri
- Department of Pharmacology and Neuroscience, Creighton University School of Medicine, Omaha, NE 68178, USA
| | - Yoland Smith
- Yerkes National Primate Research Center, Atlanta, GA 30329, USA; UDALL Center of Excellence for Parkinson's Disease, Atlanta, GA 30329, USA; Dept. Neurology, Emory University, Atlanta, GA 30329, USA
| | - Shashank M Dravid
- Department of Pharmacology and Neuroscience, Creighton University School of Medicine, Omaha, NE 68178, USA.
| |
Collapse
|
31
|
Schaffler MD, Middleton LJ, Abdus-Saboor I. Mechanisms of Tactile Sensory Phenotypes in Autism: Current Understanding and Future Directions for Research. Curr Psychiatry Rep 2019; 21:134. [PMID: 31807945 PMCID: PMC6900204 DOI: 10.1007/s11920-019-1122-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
PURPOSE OF REVIEW This review aims to summarize the current body of behavioral, physiological, and molecular knowledge concerning tactile sensitivity in autism spectrum disorder (ASD), with a focus on recent studies utilizing rodent models. RECENT FINDINGS Mice with mutations in the ASD-related genes, Shank3, Fmr1, UBE3A, and Mecp2, display tactile abnormalities. Some of these abnormalities appear to be caused by mutation-related changes in the PNS, as opposed to changes in the processing of touch stimuli in the CNS, as previously thought. There is also growing evidence suggesting that peripheral mechanisms may contribute to some of the core symptoms and common comorbidities of ASD. Researchers are therefore beginning to assess the therapeutic potential of targeting the PNS in treating some of the core symptoms of ASD. Sensory abnormalities are common in rodent models of ASD. There is growing evidence that sensory hypersensitivity, especially tactile sensitivity, may contribute to social deficits and other autism-related behaviors.
Collapse
Affiliation(s)
- Melanie D Schaffler
- Department of Biology, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Leah J Middleton
- Department of Biology, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Ishmail Abdus-Saboor
- Department of Biology, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
32
|
Nakamoto C, Konno K, Miyazaki T, Nakatsukasa E, Natsume R, Abe M, Kawamura M, Fukazawa Y, Shigemoto R, Yamasaki M, Sakimura K, Watanabe M. Expression mapping, quantification, and complex formation of GluD1 and GluD2 glutamate receptors in adult mouse brain. J Comp Neurol 2019; 528:1003-1027. [DOI: 10.1002/cne.24792] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 09/04/2019] [Accepted: 09/19/2019] [Indexed: 01/24/2023]
Affiliation(s)
- Chihiro Nakamoto
- Department of Animal Model Development Brain Research Institute, Niigata University Niigata Japan
| | - Kohtarou Konno
- Department of Anatomy, Faculty of Medicine Hokkaido University Sapporo Japan
| | - Taisuke Miyazaki
- Department of Anatomy, Faculty of Medicine Hokkaido University Sapporo Japan
- Department of Functioning and Disability, Faculty of Health Sciences Hokkaido University Sapporo Japan
| | - Ena Nakatsukasa
- Department of Animal Model Development Brain Research Institute, Niigata University Niigata Japan
| | - Rie Natsume
- Department of Animal Model Development Brain Research Institute, Niigata University Niigata Japan
| | - Manabu Abe
- Department of Animal Model Development Brain Research Institute, Niigata University Niigata Japan
| | - Meiko Kawamura
- Department of Animal Model Development Brain Research Institute, Niigata University Niigata Japan
| | - Yugo Fukazawa
- Division of Brain Structure and Function, Research Center for Child Mental Development, Life Science Advancement Program, Faculty of Medical Science University of Fukui Fukui Japan
| | - Ryuichi Shigemoto
- Institute of Science and Technology (IST Austria) Klosterneuburg Austria
| | - Miwako Yamasaki
- Department of Anatomy, Faculty of Medicine Hokkaido University Sapporo Japan
| | - Kenji Sakimura
- Department of Animal Model Development Brain Research Institute, Niigata University Niigata Japan
| | - Masahiko Watanabe
- Department of Anatomy, Faculty of Medicine Hokkaido University Sapporo Japan
| |
Collapse
|
33
|
Dohaku R, Yamaguchi M, Yamamoto N, Shimizu T, Osakada F, Hibi M. Tracing of Afferent Connections in the Zebrafish Cerebellum Using Recombinant Rabies Virus. Front Neural Circuits 2019; 13:30. [PMID: 31068795 PMCID: PMC6491863 DOI: 10.3389/fncir.2019.00030] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Accepted: 04/10/2019] [Indexed: 12/31/2022] Open
Abstract
The cerebellum is involved in some forms of motor coordination and learning, and in cognitive and emotional functions. To elucidate the functions of the cerebellum, it is important to unravel the detailed connections of the cerebellar neurons. Although the cerebellar neural circuit structure is generally conserved among vertebrates, it is not clear whether the cerebellum receives and processes the same or similar information in different vertebrate species. Here, we performed monosynaptic retrograde tracing with recombinant rabies viruses (RV) to identify the afferent connections of the zebrafish cerebellar neurons. We used a G-deleted RV that expressed GFP. The virus was also pseudotyped with EnvA, an envelope protein of avian sarcoma and leucosis virus (ALSV-A). For the specific infection of cerebellar neurons, we expressed the RV glycoprotein (G) gene and the envelope protein TVA, which is the receptor for EnvA, in Purkinje cells (PCs) or granule cells (GCs), using the promoter for aldolase Ca (aldoca) or cerebellin 12 (cbln12), respectively. When the virus infected PCs in the aldoca line, GFP was detected in the PCs’ presynaptic neurons, including GCs and neurons in the inferior olivary nuclei (IOs), which send climbing fibers (CFs). These observations validated the RV tracing method in zebrafish. When the virus infected GCs in the cbln12 line, GFP was again detected in their presynaptic neurons, including neurons in the pretectal nuclei, the nucleus lateralis valvulae (NLV), the central gray (CG), the medial octavolateralis nucleus (MON), and the descending octaval nucleus (DON). GFP was not observed in these neurons when the virus infected PCs in the aldoca line. These precerebellar neurons generally agree with those reported for other teleost species and are at least partly conserved with those in mammals. Our results demonstrate that the RV system can be used for connectome analyses in zebrafish, and provide fundamental information about the cerebellar neural circuits, which will be valuable for elucidating the functions of cerebellar neural circuits in zebrafish.
Collapse
Affiliation(s)
- Ryuji Dohaku
- Division of Biological Science, Graduate School of Science, Nagoya University, Nagoya, Japan
| | - Masahiro Yamaguchi
- Laboratory of Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya University, Nagoya, Japan
| | - Naoyuki Yamamoto
- Department of Animal Sciences, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Japan
| | - Takashi Shimizu
- Division of Biological Science, Graduate School of Science, Nagoya University, Nagoya, Japan.,Laboratory of Organogenesis and Organ Function, Bioscience and Biotechnology, Nagoya University, Nagoya, Japan
| | - Fumitaka Osakada
- Laboratory of Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya University, Nagoya, Japan
| | - Masahiko Hibi
- Division of Biological Science, Graduate School of Science, Nagoya University, Nagoya, Japan.,Laboratory of Organogenesis and Organ Function, Bioscience and Biotechnology, Nagoya University, Nagoya, Japan
| |
Collapse
|
34
|
Altered Glutamate Receptor Ionotropic Delta Subunit 2 Expression in Stau2-Deficient Cerebellar Purkinje Cells in the Adult Brain. Int J Mol Sci 2019; 20:ijms20071797. [PMID: 30979012 PMCID: PMC6480955 DOI: 10.3390/ijms20071797] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 04/02/2019] [Accepted: 04/08/2019] [Indexed: 01/13/2023] Open
Abstract
Staufen2 (Stau2) is an RNA-binding protein that is involved in dendritic spine morphogenesis and function. Several studies have recently investigated the role of Stau2 in the regulation of its neuronal target mRNAs, with particular focus on the hippocampus. Here, we provide evidence for Stau2 expression and function in cerebellar Purkinje cells. We show that Stau2 downregulation (Stau2GT) led to an increase of glutamate receptor ionotropic delta subunit 2 (GluD2) in Purkinje cells when animals performed physical activity by voluntary wheel running compared with the age-matched wildtype (WT) mice (C57Bl/6J). Furthermore, Stau2GT mice showed lower performance in motor coordination assays but enhanced motor learning abilities than did WT mice, concomitantly with an increase in dendritic GluD2 expression. Together, our results suggest the novel role of Stau2 in Purkinje cell synaptogenesis in the mouse cerebellum.
Collapse
|
35
|
Conditional knockout of UBC13 produces disturbances in gait and spontaneous locomotion and exploration in mice. Sci Rep 2019; 9:4379. [PMID: 30867488 PMCID: PMC6416404 DOI: 10.1038/s41598-019-40714-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 02/19/2019] [Indexed: 11/26/2022] Open
Abstract
Here we have characterized the functional impairments resulting from conditional knockout of the ubiquitin-conjugating E2 enzyme (UBC13) in rodent cerebellar granule neurons, which greatly increases the parallel fiber presynaptic boutons and functional parallel fiber/Purkinje cell synapses. We report that conditional UBC13 knockout mice exhibit reliable deficits on several gait-related variables when their velocity of ambulation is tightly controlled by a moving treadmill and by restricting space for movement. Selected gait parameters and movement patterns related to spontaneous exploration in an open field may also be affected in conditional UBC13 knockout mice. Analysis of open-field data as a function of test session half using force-plate actometer instrumentation suggest that conditional UBC13 knockout mice have alterations in emotionality, possibly affecting gait and movement variables. These findings suggest that conditional UBC13 knockout mice represent a valuable platform for assessing the effects of disturbances in cerebellar granule cell circuitry on gait and other aspects of locomotion. Also, the possibility that psychological factors such as altered emotionality may impact gait and movement patterns in these mice suggest that these mice may provide a useful model for evaluating analogous behavioral impairments in autism spectrum disorders and other neurodevelopmental syndromes associated with deregulation of ubiquitin signaling.
Collapse
|
36
|
Shipman ML, Green JT. Cerebellum and cognition: Does the rodent cerebellum participate in cognitive functions? Neurobiol Learn Mem 2019; 170:106996. [PMID: 30771461 DOI: 10.1016/j.nlm.2019.02.006] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 01/02/2019] [Accepted: 02/08/2019] [Indexed: 02/06/2023]
Abstract
There is a widespread, nearly complete consensus that the human and non-human primate cerebellum is engaged in non-motor, cognitive functions. This body of research has implicated the lateral portions of lobule VII (Crus I and Crus II) and the ventrolateral dentate nucleus. With rodents, however, it is not so clear. We review here approximately 40 years of experiments using a variety of cerebellar manipulations in rats and mice and measuring the effects on executive functions (working memory, inhibition, and cognitive flexibility), spatial navigation, discrimination learning, and goal-directed and stimulus-driven instrumental conditioning. Our conclusion is that there is a solid body of support for engagement of the rodent cerebellum in tests of cognitive flexibility and spatial navigation, and some support for engagement in working memory and certain types of discrimination learning. Future directions will involve determining the relevant cellular mechanisms, cerebellar regions, and precise cognitive functions of the rodent cerebellum.
Collapse
Affiliation(s)
- Megan L Shipman
- Department of Psychological Science, University of Vermont, 2 Colchester Avenue, Burlington, VT 05405, USA; Neuroscience Graduate Program, University of Vermont, 2 Colchester Avenue, Burlington, VT 05405, USA.
| | - John T Green
- Department of Psychological Science, University of Vermont, 2 Colchester Avenue, Burlington, VT 05405, USA.
| |
Collapse
|
37
|
A central amygdala to zona incerta projection is required for acquisition and remote recall of conditioned fear memory. Nat Neurosci 2018; 21:1515-1519. [PMID: 30349111 DOI: 10.1038/s41593-018-0248-4] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Accepted: 08/28/2018] [Indexed: 11/08/2022]
Abstract
The formation and retrieval of conditioned fear memories critically depend on the amygdala. Here we identify an inhibitory projection from somatostatin-positive neurons in the central amygdala to parvalbumin-positive neurons in the zona incerta that is required for both recent and remote fear memories. Thus, the amygdala inhibitory input to parvalbumin-positive neurons in the zona incerta, a nucleus not previously implicated in fear memory, is an essential component of the fear memory circuitry.
Collapse
|
38
|
Cbln2 and Cbln4 are expressed in distinct medial habenula-interpeduncular projections and contribute to different behavioral outputs. Proc Natl Acad Sci U S A 2018; 115:E10235-E10244. [PMID: 30287486 DOI: 10.1073/pnas.1811086115] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Cerebellins are important neurexin ligands that remain incompletely understood. Two critical questions in particular remain unanswered: do different cerebellins perform distinct functions, and do these functions act in the initial establishment of synapses or in rendering nascent synapses capable of normal synaptic transmission? Here we show that in mice, Cbln2 and Cbln4 are expressed in the medial habenula (MHb) nucleus in different types of neurons that project to distinct target neurons in the interpeduncular nucleus. Conditional genetic deletion of Cbln2 in the MHb impaired synaptic transmission at Cbln2+ synapses in the interpeduncular neurons within 3 wk, but decreased synapse numbers only after 3 mo, suggesting a functional, but not a structural, requirement for Cbln2 in synapses formed by Cbln2-expressing neurons. In contrast, genetic deletions of Cbln4 in the MHb had no major effect on synaptic transmission or synapse numbers in interpeduncular target neurons. Nevertheless, MHb ablation of both Cbln2 and Cbln4 significantly impaired behavioral responses in mice, but affected different types of behaviors. Specifically, Cbln2 MHb deletions decreased spatial learning, as measured in the water T-maze, whereas Cbln4 MHb deletions increased anxiety levels, as monitored in the open field test and elevated plus maze. Thus, Cbln2 and Cbln4 are expressed in distinct MHb neurons that contribute to different behaviors.
Collapse
|
39
|
Moreno-Rius J. The cerebellum in fear and anxiety-related disorders. Prog Neuropsychopharmacol Biol Psychiatry 2018; 85:23-32. [PMID: 29627508 DOI: 10.1016/j.pnpbp.2018.04.002] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2017] [Revised: 03/29/2018] [Accepted: 04/04/2018] [Indexed: 01/06/2023]
Abstract
Fear and anxiety-related disorders are highly prevalent psychiatric conditions characterized by avoidant and fearful reactions towards specific stimuli or situations, which are disproportionate given the real threat such stimuli entail. These conditions comprise the most common mental disorder group. There are a high proportion of patients who fail to achieve remission and the presence of high relapse rates indicate the therapeutic options available are far from being fully efficient. Despite an increased understanding the neural circuits underlying fear and anxiety-related behaviors in the last decades, a factor that could be partially contributing to the lack of adequate therapies may be an insufficient understanding of the core features of the disorders and their associated neurobiology. Interestingly, the cerebellum shows connections with fear and anxiety-related brain areas and functional involvement in such processes, but explanations for its role in anxiety disorders are lacking. Therefore, the aims of this review are to provide an overview of the neural circuitry of fear and anxiety and its connections to the cerebellum, and of the animal studies that directly assess an involvement of the cerebellum in these processes. Then, the studies performed in patients suffering from anxiety disorders that explore the cerebellum will be discussed. Finally, we'll propose a function for the cerebellum in these disorders, which could guide future experimental approaches to the topic and lead to a better understanding of the neurobiology of anxiety-related disorders, ultimately helping to develop more effective treatments for these conditions.
Collapse
Affiliation(s)
- Josep Moreno-Rius
- Department of Pharmacology and Toxicology, University of Innsbruck, Innsbruck, Austria.
| |
Collapse
|
40
|
Park D, Bae S, Yoon TH, Ko J. Molecular Mechanisms of Synaptic Specificity: Spotlight on Hippocampal and Cerebellar Synapse Organizers. Mol Cells 2018; 41:373-380. [PMID: 29665671 PMCID: PMC5974614 DOI: 10.14348/molcells.2018.0081] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2018] [Revised: 03/30/2018] [Accepted: 04/02/2018] [Indexed: 12/13/2022] Open
Abstract
Synapses and neural circuits form with exquisite specificity during brain development to allow the precise and appropriate flow of neural information. Although this property of synapses and neural circuits has been extensively investigated for more than a century, molecular mechanisms underlying this property are only recently being unveiled. Recent studies highlight several classes of cell-surface proteins as organizing hubs in building structural and functional architectures of specific synapses and neural circuits. In the present mini-review, we discuss recent findings on various synapse organizers that confer the distinct properties of specific synapse types and neural circuit architectures in mammalian brains, with a particular focus on the hippocampus and cerebellum.
Collapse
Affiliation(s)
- Dongseok Park
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988,
Korea
| | - Sungwon Bae
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988,
Korea
| | - Taek Han Yoon
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988,
Korea
| | - Jaewon Ko
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988,
Korea
| |
Collapse
|
41
|
Postsynaptic δ1 glutamate receptor assembles and maintains hippocampal synapses via Cbln2 and neurexin. Proc Natl Acad Sci U S A 2018; 115:E5373-E5381. [PMID: 29784783 DOI: 10.1073/pnas.1802737115] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The δ1 glutamate receptor (GluD1) was cloned decades ago and is widely expressed in many regions of the brain. However, its functional roles in these brain circuits remain unclear. Here, we find that GluD1 is required for both excitatory synapse formation and maintenance in the hippocampus. The action of GluD1 is absent in the Cbln2 knockout mouse. Furthermore, the GluD1 actions require the presence of presynaptic neurexin 1β carrying the splice site 4 insert (+S4). Together, our findings demonstrate that hippocampal synapse assembly and maintenance require a tripartite molecular complex in which the ligand Cbln2 binds with presynaptic neurexin 1β (+S4) and postsynaptic GluD1. We provide evidence that this mechanism may apply to other forebrain synapses, where GluD1 is widely expressed.
Collapse
|
42
|
Rong Y, Bansal PK, Wei P, Guo H, Correia K, Parris J, Morgan JI. Glycosylation of Cblns attenuates their receptor binding. Brain Res 2018; 1694:129-139. [PMID: 29782851 DOI: 10.1016/j.brainres.2018.05.022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 05/11/2018] [Accepted: 05/18/2018] [Indexed: 01/11/2023]
Abstract
Cbln1 is the prototype of a family (Cbln1-Cbln4) of secreted glycoproteins and is essential for normal synapse structure and function in cerebellum by bridging presynaptic Nrxn to postsynaptic Grid2. Here we report the effects of glycosylation on the in vitro receptor binding properties of Cblns. Cbln1, 2 and 4 harbor two N-linked glycosylation sites, one at the N-terminus is in a region implicated in Nrxn binding and the second is in the C1q domain, a region involved in Grid2 binding. Mutation (asparagine to glutamine) of the N-terminal site, increased neurexin binding whereas mutation of the C1q site markedly increased Grid2 binding. These mutations did not influence subunit composition of Cbln trimeric complexes (mediated through the C1q domain) nor their assembly into hexamers (mediated by the N-terminal region). Therefore, glycosylation likely masks the receptor binding interfaces of Cblns. As Cbln4 has undetectable Grid2 binding in vitro we assessed whether transgenic expression of wild type Cbln4 or its glycosylation mutants rescued the Cbln1-null phenotype in vivo. Cbln4 partially rescued and both glycosylation mutants completely rescued ataxia in cbln1-null mice. Thus Cbln4 has intrinsic Grid2 binding that is attenuated by glycosylation, and glycosylation mutants exhibit gain of function in vivo.
Collapse
Affiliation(s)
- Yongqi Rong
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Parmil K Bansal
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Peng Wei
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Hong Guo
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Kristen Correia
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Jennifer Parris
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - James I Morgan
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA.
| |
Collapse
|
43
|
Seigneur E, Südhof TC. Genetic Ablation of All Cerebellins Reveals Synapse Organizer Functions in Multiple Regions Throughout the Brain. J Neurosci 2018; 38:4774-4790. [PMID: 29691328 PMCID: PMC5956990 DOI: 10.1523/jneurosci.0360-18.2018] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 03/27/2018] [Accepted: 04/16/2018] [Indexed: 01/26/2023] Open
Abstract
Cerebellins are synaptic organizer molecules that bind to presynaptic neurexins and postsynaptic receptors. They are well studied in the cerebellum, but three of the four cerebellins (Cbln1, Cbln2, and Cbln4) are also broadly expressed outside of the cerebellum, suggesting that they perform general functions throughout the brain. Here, we generated male and female constitutive single (KO), double KO (dKO), and triple KO (tKO) mice of Cbln1, Cbln2, and Cbln4. We found that all constitutive cerebellin-deficient mice were viable and fertile, suggesting that cerebellins are not essential for survival. Cbln1/2 dKO mice exhibited salience-induced seizures that were aggravated in Cbln1/2/4 tKO mice, suggesting that all cerebellins contribute to brain function. As described previously, Cbln1 KO mice displayed major motor impairments that were aggravated by additional KO of Cbln2. Strikingly, the Cbln1/2 dKO did not cause alterations in synapse density in the hippocampus of young adult (1- and 2-month-old) mice, but produced a selective ∼50% decrease in hippocampal synapse density in the stratum lacunosum moleculare of the CA1 region and in the dentate gyrus of aging, 6-month-old mice. A similar decrease in excitatory synapse density was observed in the striatum and retrosplenial cortex. Behaviorally, the Cbln1 KO produced dramatic changes in motor behaviors that were partly aggravated by additional deletion of Cbln2 and/or Cbln4. Our results show that cerebellins are not essential for survival and do not contribute to initial synapse formation, but perform multiple functions throughout the brain; as a consequence, their ablation results in a delayed loss of synapses and in behavioral impairments.SIGNIFICANCE STATEMENT Cerebellins (Cbln1-4) are trans-synaptic cell adhesion molecules. In the cerebellum, Cbln1 functions as a bidirectional organizer of parallel fiber-Purkinje cell synapses by binding to presynaptic neurexins and postsynaptic GluRδ2. Little is known about the function of cerebellins outside of the cerebellum; therefore, the present study used single, double, and triple constitutive KO mice of Cbln1, Cbln2, and Cbln4 to analyze the overall function of cerebellins. We show that cerebellins act as important synaptic organizers in specific subsets of neurons and likely contribute to many different brain functions. We also show that cerebellins are not initially required for synapse formation, but rather for specification and long-term synapse maintenance and demonstrate that all cerebellins, not just Cbln1, contribute to brain function.
Collapse
Affiliation(s)
- Erica Seigneur
- Department of Molecular and Cellular Physiology and Howard Hughes Medical Institute, Stanford University Medical School, Stanford, California 94305
| | - Thomas C Südhof
- Department of Molecular and Cellular Physiology and Howard Hughes Medical Institute, Stanford University Medical School, Stanford, California 94305
| |
Collapse
|
44
|
Ferrer-Ferrer M, Dityatev A. Shaping Synapses by the Neural Extracellular Matrix. Front Neuroanat 2018; 12:40. [PMID: 29867379 PMCID: PMC5962695 DOI: 10.3389/fnana.2018.00040] [Citation(s) in RCA: 102] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 04/25/2018] [Indexed: 11/13/2022] Open
Abstract
Accumulating data support the importance of interactions between pre- and postsynaptic neuronal elements with astroglial processes and extracellular matrix (ECM) for formation and plasticity of chemical synapses, and thus validate the concept of a tetrapartite synapse. Here we outline the major mechanisms driving: (i) synaptogenesis by secreted extracellular scaffolding molecules, like thrombospondins (TSPs), neuronal pentraxins (NPs) and cerebellins, which respectively promote presynaptic, postsynaptic differentiation or both; (ii) maturation of synapses via reelin and integrin ligands-mediated signaling; and (iii) regulation of synaptic plasticity by ECM-dependent control of induction and consolidation of new synaptic configurations. Particularly, we focused on potential importance of activity-dependent concerted activation of multiple extracellular proteases, such as ADAMTS4/5/15, MMP9 and neurotrypsin, for permissive and instructive events in synaptic remodeling through localized degradation of perisynaptic ECM and generation of proteolytic fragments as inducers of synaptic plasticity.
Collapse
Affiliation(s)
- Maura Ferrer-Ferrer
- Molecular Neuroplasticity German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
| | - Alexander Dityatev
- Molecular Neuroplasticity German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany.,Center for Behavioral Brain Sciences (CBBS), Magdeburg, Germany.,Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
| |
Collapse
|
45
|
Südhof TC. Synaptic Neurexin Complexes: A Molecular Code for the Logic of Neural Circuits. Cell 2017; 171:745-769. [PMID: 29100073 DOI: 10.1016/j.cell.2017.10.024] [Citation(s) in RCA: 501] [Impact Index Per Article: 71.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Revised: 10/04/2017] [Accepted: 10/15/2017] [Indexed: 10/18/2022]
Abstract
Synapses are specialized junctions between neurons in brain that transmit and compute information, thereby connecting neurons into millions of overlapping and interdigitated neural circuits. Here, we posit that the establishment, properties, and dynamics of synapses are governed by a molecular logic that is controlled by diverse trans-synaptic signaling molecules. Neurexins, expressed in thousands of alternatively spliced isoforms, are central components of this dynamic code. Presynaptic neurexins regulate synapse properties via differential binding to multifarious postsynaptic ligands, such as neuroligins, cerebellin/GluD complexes, and latrophilins, thereby shaping the input/output relations of their resident neural circuits. Mutations in genes encoding neurexins and their ligands are associated with diverse neuropsychiatric disorders, especially schizophrenia, autism, and Tourette syndrome. Thus, neurexins nucleate an overall trans-synaptic signaling network that controls synapse properties, which thereby determines the precise responses of synapses to spike patterns in a neuron and circuit and which is vulnerable to impairments in neuropsychiatric disorders.
Collapse
Affiliation(s)
- Thomas C Südhof
- Department of Molecular and Cellular Physiology and Howard Hughes Medical Institute, Stanford University Medical School, 265 Campus Drive, CA 94305-5453, USA.
| |
Collapse
|
46
|
Abstract
Research in the last two decades has identified many synaptic organizers in the central nervous system that directly regulate the assembly of pre- and/or postsynaptic molecules, such as synaptic vesicles, active zone proteins, and neurotransmitter receptors. They are classified into secreted factors and cell adhesion molecules, such as neurexins and neuroligins. Certain secreted factors are termed extracellular scaffolding proteins (ESPs) because they are components of the synaptic extracellular matrix and serve as a scaffold at the synaptic cleft. These include Lgi1, Cbln1, neuronal pentraxins, Hevin, thrombospondins, and glypicans. Diffusible secreted factors, such as Wnts, fibroblast growth factors, and semaphorins, tend to act from a distance. In contrast, ESPs remain at the synaptic cleft and often help synaptic adhesion and/or accumulation of postsynaptic receptors. Many fundamental questions remain about when, how, and why various synaptic organizers establish and modify the vast numbers of connections during development and throughout life.
Collapse
Affiliation(s)
- Michisuke Yuzaki
- Department of Physiology, Keio University School of Medicine, Tokyo 160-8582, Japan;
| |
Collapse
|
47
|
Yuzaki M. The C1q complement family of synaptic organizers: not just complementary. Curr Opin Neurobiol 2017; 45:9-15. [DOI: 10.1016/j.conb.2017.02.002] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Revised: 01/25/2017] [Accepted: 02/01/2017] [Indexed: 11/26/2022]
|
48
|
Seigneur E, Südhof TC. Cerebellins are differentially expressed in selective subsets of neurons throughout the brain. J Comp Neurol 2017; 525:3286-3311. [PMID: 28714144 DOI: 10.1002/cne.24278] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Revised: 06/15/2017] [Accepted: 06/27/2017] [Indexed: 12/13/2022]
Abstract
Cerebellins are secreted hexameric proteins that form tripartite complexes with the presynaptic cell-adhesion molecules neurexins or 'deleted-in-colorectal-cancer', and the postsynaptic glutamate-receptor-related proteins GluD1 and GluD2. These tripartite complexes are thought to regulate synapses. However, cerebellins are expressed in multiple isoforms whose relative distributions and overall functions are not understood. Three of the four cerebellins, Cbln1, Cbln2, and Cbln4, autonomously assemble into homohexamers, whereas the Cbln3 requires Cbln1 for assembly and secretion. Here, we show that Cbln1, Cbln2, and Cbln4 are abundantly expressed in nearly all brain regions, but exhibit strikingly different expression patterns and developmental dynamics. Using newly generated knockin reporter mice for Cbln2 and Cbln4, we find that Cbln2 and Cbln4 are not universally expressed in all neurons, but only in specific subsets of neurons. For example, Cbln2 and Cbln4 are broadly expressed in largely non-overlapping subpopulations of excitatory cortical neurons, but only sparse expression was observed in excitatory hippocampal neurons of the CA1- or CA3-region. Similarly, Cbln2 and Cbln4 are selectively expressed, respectively, in inhibitory interneurons and excitatory mitral projection neurons of the main olfactory bulb; here, these two classes of neurons form dendrodendritic reciprocal synapses with each other. A few brain regions, such as the nucleus of the lateral olfactory tract, exhibit astoundingly high Cbln2 expression levels. Viewed together, our data show that cerebellins are abundantly expressed in relatively small subsets of neurons, suggesting specific roles restricted to subsets of synapses.
Collapse
Affiliation(s)
- Erica Seigneur
- Department of Molecular & Cellular Physiology and Howard Hughes Medical Institute, Stanford University Medical School, Stanford, California
| | - Thomas C Südhof
- Department of Molecular & Cellular Physiology and Howard Hughes Medical Institute, Stanford University Medical School, Stanford, California
| |
Collapse
|
49
|
Yuzaki M, Aricescu AR. A GluD Coming-Of-Age Story. Trends Neurosci 2017; 40:138-150. [PMID: 28110935 DOI: 10.1016/j.tins.2016.12.004] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Revised: 12/19/2016] [Accepted: 12/22/2016] [Indexed: 01/02/2023]
Abstract
The GluD1 and GluD2 receptors form the GluD ionotropic glutamate receptor (iGluR) subfamily. Without known endogenous ligands, they have long been referred to as 'orphan' and remained enigmatic functionally. Recent progress has, however, radically changed this view. Both GluD receptors are expressed in wider brain regions than originally thought. Human genetic studies and analyses of knockout mice have revealed their involvement in multiple neurodevelopmental and psychiatric disorders. The discovery of endogenous ligands, together with structural investigations, has opened the way towards a mechanistic understanding of GluD signaling at central nervous system synapses. These studies have also prompted the hypothesis that all iGluRs, and potentially other neurotransmitter receptors, rely on the cooperative binding of extracellular small-molecule and protein ligands for physiological signaling.
Collapse
Affiliation(s)
- Michisuke Yuzaki
- Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan.
| | - A Radu Aricescu
- Division of Structural Biology, Wellcome Trust Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
| |
Collapse
|